EP2333067A1 - Method for isolation of soluble polypeptides - Google Patents

Method for isolation of soluble polypeptides Download PDF

Info

Publication number
EP2333067A1
EP2333067A1 EP10190246A EP10190246A EP2333067A1 EP 2333067 A1 EP2333067 A1 EP 2333067A1 EP 10190246 A EP10190246 A EP 10190246A EP 10190246 A EP10190246 A EP 10190246A EP 2333067 A1 EP2333067 A1 EP 2333067A1
Authority
EP
European Patent Office
Prior art keywords
antibody fragment
protein
library
human
sequence
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP10190246A
Other languages
German (de)
French (fr)
Inventor
Jamshid Tanha
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
National Research Council of Canada
Original Assignee
National Research Council of Canada
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by National Research Council of Canada filed Critical National Research Council of Canada
Publication of EP2333067A1 publication Critical patent/EP2333067A1/en
Withdrawn legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/005Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies constructed by phage libraries
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/02Drugs for genital or sexual disorders; Contraceptives for disorders of the vagina
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1267Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria
    • C07K16/1271Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria from Micrococcaceae (F), e.g. Staphylococcus
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1267Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria
    • C07K16/1282Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria from Clostridium (G)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1037Screening libraries presented on the surface of microorganisms, e.g. phage display, E. coli display
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • G01N33/6845Methods of identifying protein-protein interactions in protein mixtures
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance

Definitions

  • This invention relates to the isolation, identification and manipulation of polypeptides, especially monomeric human antibody fragments.
  • Antibodies in vertebrates are typically composed of paired heavy (H) and light (L) chains.
  • the first domain of the combined H and L chains, the V H and V L are more variable in sequence, and this is the portion of the antibody that recognizes and binds to the antigen.
  • the V H and V L domains recognize the antigen as a pair.
  • V H H The immune repertoire of camelidae (camels, dromedaries and llamas) is unique in that it possesses unusual types of antibodies referred to as heavy-chain antibodies (Hamers, Casterman C. et al., 1993). These antibodies lack light chains and thus their combining sites consist of one domain, termed V H H.
  • V H H single-domain antibodies provide several advantages over single-chain Fv (scFv) fragments derived from conventional four-chain antibodies. While sdAbs are comparable to their scFv counterparts in terms of affinity, they outperform scFvs in terms of solubility, stability, resistance to aggregation, refoldability, expression yield, and ease of DNA manipulation, library construction and 3-D structural determinations. Many of the aforementioned properties of V H H sdAbs are desired in applications involving antibodies.
  • V H Hs limits their use in human immunotherapy due to immunogenicity.
  • human V H and V L sdAbs are ideal candidates for immunotherapy applications because they are expected to be least immunogenic.
  • V H s and V L s are by and large prone to aggregation, a characteristic common to V H s and V L s derived from conventional antibodies (Davies, J. et al., 1994;Tanha, J. et al., 2001;Ward, E. S. et al., 1989). Thus, attempts have been made to obtain monomer human V H s and V L s suitable for antibody applications. Such V H s and V L s have also displayed other useful properties typical of V H Hs such as high expression yield, high refoldability and resistance to aggregation. Synthetic libraries built on these V H s and V L s as library scaffolds might serve as a promising source of therapeutic proteins.
  • V H s and V L s were isolated from human synthetic V H and V L libraries without resorting to engineering of the sort mentioned above.
  • a monomeric human V H was discovered when a human V H library was panned against hen egg lysozyme (Jespers, L. et al., 2004b).
  • a selection method based on reversible unfolding and affinity criteria yielded many monomeric V H s from synthetic human V H libraries (Jespers, L. et al., 2004a). This finding underlined the fact that an appropriate selection method is key to efficient capturing of rare monomer human V H s with desirable biophysical properties.
  • a first object of the invention is to provide a high throughput screening method for identifying polypeptides, especially antibody fragments, with improved biophysical properties, including solubility, high expression, and/or stability (such as high refolding after thermal denaturation, high resistance to chemical denaturant, and high resistance to proteases, in particular gastrointestinal proteases such as trypsin).
  • a second object of the invention is to provide a high throughput screening method for identifying monomeric human V H s and V L s.
  • a third object of the invention is to identify, isolate and characterize monomeric human V H s and V L s.
  • a fourth object of the invention is to construct and characterize multimers of antibody fragments, especially monomeric human V H s and V L s.
  • a fifth object of the invention is to construct display libraries from polypeptides, especially antibody fragments, and most especially monomeric human V H s and V L s.
  • a sixth object of the invention is to provide a DNA shuffling method for producing polypeptides, especially antibody fragments, and most especially monomeric human V H s and V L s with improved biophysical properties.
  • the method includes the steps of obtaining a phage display library capable of expressing a variety of polypeptide sequences, allowing infection of a bacterial lawn by the library phage, and identifying phage which form larger than average plaques on the bacterial lawn. The phage are then isolated, and steps are taken to sequence or otherwise characterize the polypeptide sequences.
  • the invention also provides for polypeptides, especially monomeric human V H s and V L s, identified by the above method, which may be useful for immunotherapy, and/or as diagnostic or detection agents.
  • the monomeric human V H s and V L s may also be combined to form dimers, trimers, pentamers or other multimers, which may be useful for immunotherapy and/or as diagnostic or detection agents.
  • polypeptides identified by the above method can be manipulated by methods such as DNA shuffling to select for improved biophysical properties such as solubility, stability, monomericity, high expressibility, binding specificity and human origin.
  • polypeptides identified by the above method including human V H s and V L s, may also be used to generate further display libraries, which can then in turn be used to isolate further polypeptides by the above method.
  • the present invention provides a method of identifying target polypeptides , comprising a) obtaining a phage display library capable of expressing a variety of polypeptide sequences, b) allowing infection of a bacterial lawn by the library phage and c) identifying phage which form larger than average plaques on the bacterial lawn.
  • the present invention provides polypeptide having an amino acid sequence selected from the group consisting of: SEQ ID NO:8-54
  • the present invention provides a V H antibody fragment comprising at least one amino acid sequence selected from the group consisting of: SEQ ID NO:8-22.
  • the present invention provides a V L antibody fragment comprising at least one amino acid sequence selected from the group consisting of: SEQ ID NO:23-54.
  • the present invention provides A method for producing polypeptides with desirable biophysical properties, comprising the steps of a) providing at least one first nucleic acid sequence that encodes an antibody fragment as claimed in claim 41,42,44,45,47,48, 59 or 70 or that encodes a polypeptide sequence as claimed in claim 24,27,37 or 39, and having a first desirable property; b) providing at least one second nucleic acid sequence that encodes an antibody fragment having a second desirable property; c) cutting the at least one first and at least one second nucleic acid sequences into random fragments; d) reassembling the random fragments; e) expressing the random fragments; and f) screening the expressed random fragments for the first and second desirable properties.
  • polypeptides especially antibody fragments, that are of human origin, soluble, stable, resistant to aggregation, refoldable, highly expressed, easily manipulated at the DNA level, ideal for library construction and for 3-D structural determinations.
  • antibody fragments are useful for a wide variety of immunotherapeutical applications, and also as diagnostic and detection agents.
  • Human monomeric V H and V L antibodies are of particular interest, as they are likely to have many of the above-mentioned properties.
  • Polypeptides with the above-mentioned properties may be identified by high throughput screening of libraries capable of expressing a variety of polypeptide sequences.
  • phage display libraries preferably filamentous phage such as M13 or fd
  • phage display libraries may be screened by infecting a field of bacteria susceptible to the phage (a bacterial lawn) with the phage, then determining which phages have successfully lysed the bacteria by looking for clear, bacteria-free areas known as plaques.
  • Phages displaying monomeric Ilaminated V H s and V L s form larger plaques on bacterial lawns than phages displaying fully human V H s with aggregation tendencies.
  • plaque size may be used as a means of identifying rare, naturally-occurring monomer V H s and V L sfrom the human V H repertoire.
  • the method disclosed herein is also useful in identifying soluble, stable (stability covers a number of characteristics, including but not limited to high thermal refolding efficiency, high melting temperature, maintaining functionality after long (several days) incubation at 37°C, resistant to chemical denaturants, resistant to proteases, having a long shelf life at below 0°C, and 4°C, and at room temperature, maintaining functionality in intracellular environments, and maintaining functionality inside the human body, such as in the bloodstream) and high expressing proteins of differing origins, including:
  • polypeptides Once polypeptides have been identified by this method, they can be used to construct additional libraries. This is done by selecting a nucleic acid sequence of, for example, a VH. Oligonucleotides with randomized codons are created and incorporated into the VH sequence. Thus, each unique oligonucleotide is incorporated into a VH gene, and the modified VH genes constitute a library of sequences with slight variations. Typically, the oligonucleotides are designed such that the CDRs or loops of the VH are randomized. For example, one, two or all three of VH CDRs may be randomized.
  • the VH library is then cloned into an appropriate vector, depending on the type of library to be used, and the nucleic acid sequences are expressed as polypeptides.
  • the library is screened for molecules that bind to the library polypeptides, typically by panning.
  • the libraries may be phage display libraries, or other display libraries such as ribosome display and yeast display.
  • Polypeptides identified by the method discussed herein may be used for immunotherapy by, for example, the cross-linking of monomers to form dimers, trimers, pentamers and other multimers. This may result in better affinity for antigen molecules and slower dissociation rates for some antigens.
  • Another possible approach is to link or fuse polypeptides to a variety of molecules with various functions. For example, antibody fragments may be linked to radionuclides, cytotoxic drugs, toxins, peptides, proteins, enzymes, liposomes, lipids, T-cell superantigens or viruses in order to target and destroy or modify specific cells or molecules.
  • V H s or V L s identified by the selection method described herein can be further manipulated to select for improved biophysical properties such as solubility, stability, monomericity, binding specificity, human origin or high expressability.
  • This can be achieved by in vitro recombination techniques such as DNA shuffling or a staggered extension process.
  • DNA shuffling involves cutting the nucleic acid sequence of first (donor) and second (acceptor) polypeptides, such as antibody fragments, into random fragments, then reassembling the random fragments by a PCR-like reaction. The reassembled fragments are then screened to select for the desired properties.
  • one or more VHs with high stability can be mixed with one or more VHs lacking sufficient stability (acceptors) and subjected to DNA shuffling.
  • This generates mutants of the acceptor VHs which have incorporated stability residues from the donor VHs.
  • the newly stable mutants can be identified by the methods described herein, or through other evolutionary protein screening systems such as ribosome display, yeast display, bacterial cell display and phage display.
  • this technique can be used to transfer desirable traits such as solubility, monomericity, and high expression.
  • This technique may be used where both donor and acceptor V H s have desirable properties, to produce a V H with both properties.
  • an unstable donor V H which binds to an important therapeutic or diagnostic ligand can be shuffled with a stable acceptor V H .
  • the screening system may involve a ligand binding step.
  • DNA shuffling may also be useful for humanizing non-human V H s such as camelid heavy chain antibody variable domains and nurse shark and wobbegong shark variable domains, or non-human V L s which bind to therapeutic targets.
  • Human V H s and V L s with desirable properties such as solubility, stability, monomericity and high expressability may be used as donors.
  • one or more human V H s with good stability can be mixed with one or more non-human therapeutic V H s (acceptors) and subjected to DNA shuffling. This generates mutants of the acceptor V H s which are both stable and humanized.
  • the newly generated humanized and stable mutants can be identified by the methods described herein, or through other evolutionary protein screening systems such as ribosome display, yeast display, bacterial cell display and phage display.
  • the acceptor V H could be a therapeutic V H H (camelid heavy chain antibody variable domain).
  • the donor polypeptide and the acceptor polypeptide may be both human, or the donor may be human and the acceptor non-human.
  • a possible approach for imparting solubility, monomericity, high expressability or stability to V H s and V L s may be through grafting complementarity determining regions (CDRs) onto acceptor V H s and V L s. Since CDRs are known to be involved in the solubility and stability of single-domain antibodies, and accordingly the grafting of these regions, such as the CDRs from V H s and V L s isolated by the methods described herein, may impart solubility and/or stability to acceptor V H s and V L s.
  • CDRs complementarity determining regions
  • V H s monomeric human V H s with different germline and overall sequences were identified (see Figure 1 and SEQ ID NO.8 through 22) from a na ⁇ ve human V H phage display library by this selection method based on phage plaque size.
  • the V H s remain functional and monomeric following trypsin treatment at 37°C, weeks of incubations at 37°C or months of storage at 4°C, have high thermal refolding efficiencies, are produced in good yields in E. coli and possess protein A binding activity.
  • V L s monomeric human V L s were identified (see Figure 6 and SEQ ID NO. 23 through 54). The V L s are also produced in good yields in E. coli and possess protein L binding activity.
  • V H s Such properties will also be manifested by V H s from synthetic libraries that utilize the above V H s as scaffolds.
  • libraries may yield therapeutic or diagnostic V H s which would have good efficacy at physiological temperature, extended shelf life and a cost-effective production.
  • High thermal refolding efficiency characteristic would further extend the biotechnological applications of these libraries to situations where V H binders are required to maintain their activity after exposure to transient high temperatures.
  • the V H s should also be very suitable for intrabody applications because of their desirable biophysical properties.
  • the protein A binding property will simplify V H purification and detection in diagnostic tests, immunoblotting and immunocytochemistry and can be exploited to enhance library performance by removing nonfunctional V H s from the libraries.
  • libraries that utilize V L s as scaffolds will yield therapeutic or diagnostic V L S which have similarly desirable properties. Since V L s bind with protein L, V L purification and detection is simplified by taking advantage of this protein L binding property.
  • V H s and V L s may also be a useful source of diagnostics and detection agents.
  • V H s In terms of library construction, it would be desirable that the monomericity of the present V H s not be dependent on CDRs, in particular CDR3, so that CDR randomization be performed without the worry of jeopardizing library stability.
  • the V H s with smaller CDR3, e.g, HVHB82 may be preferred scaffolds since there would be less dependence on CDR3 for stability.
  • V H s and V L s in terms of overall sequence and CDR3 length should allow the construction of better-performing libraries.
  • Synthetic V H libraries have been constructed on single scaffolds. Such an approach to repertoire generation is in sharp contrast to the natural, in vivo "approach" which utilizes a multiplicity of scaffolds. Based on the sequences reported here one can take advantage of the availability of the diverse set of V H s and V L s and create libraries which are based on multiple V H and V L scaffolds. Such libraries would be a better emulation of in vivo repertoires and therefore, would have a more optimal complexity.
  • CDR3 generally contribute most significantly to repertoire diversity and for this reason CDR3 randomization on V H and V L scaffolds are typically accompanied by concomitant varying of CDR3 length. While this significantly improves library complexity, it may also compromise library stability by disrupting the length of the parental scaffold CDR3.
  • the heterogeneity of the V H s and V L s disclosed herein in terms of CDR3 length permit the creation of libraries with both good complexity, good stability and good biophysical characteristics.
  • Such libraries would preferably consist of sub-libraries, where each sub-library is created by CDR3 randomization (and CDR1 and/or CDR2 randomization, if desired) on a single V H or V L scaffold without disrupting the parental CDR3 length.
  • V H s and V L s are also beneficial in terms of choosing an optimal V H or V L framework for humanizing V H Hs, V H s and V L s which are specific to therapeutic targets.
  • High affinity camelid V H Hs against therapeutic targets can be obtained from immune, non-immunized or synthetic V H H libraries with relative ease and be subsequently subjected to humanization (CDR grafting, resurfacing, deimmunization) to remove possible V H H immunogenicity, hence providing an alternative to human V H library approach for production of therapeutic V H s.
  • Generating high affinity therapeutic V H s by the latter approach may often require additional tedious and time consuming in vitro affinity maturation of the lead binder(s) selected from the primary synthetic human V H libraries.
  • Nonhuman V H s against therapeutic targets can be obtained from immune, non-immunized or synthetic V H libraries with relative ease and be subsequently subjected to humanization (CDR grafting, resurfacing, deimmunization) to eliminate nonhuman V H immunogenicity, hence providing an alternative to human V H library approach for production of therapeutic V H s.
  • Nonhuman V L s against therapeutic targets can be obtained from immune, non-immunized or synthetic V H H libraries with relative ease and be subsequently subjected to humanization (CDR grafting, resurfacing, deimmunization) to eliminate V H H immunogenicity, hence providing an alternative to human V L library approach for production of therapeutic V L s.
  • Examples of evolutionary selection approaches involving phage display include conventional phage display, selectively infective phage and the proteolysis approaches.
  • affinity selection is used to select stable species from a library, based on the assumption that stable proteins possess better binding properties for their ligand than the unstable ones.
  • these approaches may primarily enrich for higher affinity rather than for higher stability (Jung, S . et al., 1999).
  • a binding step requirement also limits the applicability of these approaches to proteins with known ligands.
  • the third, proteolysis approach is based on the fact that stable proteins are generally compact and therefore are resistant to proteases whereas the unstable ones are not.
  • the phage display format is engineered in such a way that the protease stability of the displayed protein translates to phage infectivity.
  • a variant phage display library is treated with a protease, only the phages displaying stable proteins retain their infectivity and can subsequently be selected by infecting an E. coli host. Since this approach is independent of ligand binding, it has general utility. However, even stable and well folded proteins have protease sensitive sites, e.g., loops and linkers, and this could sometimes hinder the selection of stable species in a proteolysis approach (Bai, Y. et al., 2004) .
  • proteins with superior biophysical properties are simply identified by the naked eye.
  • the approach does not require ligand binding, proteolysis or destabilization steps, and thus, avoids complications which may be encountered in the reported selection approaches.
  • No requirement for a binding step also means that this approach has general utility.
  • a binding step may be included to ensure that the selected proteins are functional.
  • the dependency of the present approach on plating introduces a possible logistical limitation in terms of the number of plates that can be handled and thus limits its application to smaller libraries. Nonetheless, the utility of the current approach can be extended to large libraries, if the library is first reduced to a manageable size.
  • V H s and V L s of good biophysical properties in a background of very unstable V H s and V L s.
  • the lead variants may be identified based on the rate of plaque formation by using shorter incubation times, or based on plaque size and frequency criteria.
  • the present selection approach can be extended to identification of stable and well-folded antibody fragments such as scFvs and Fabs with the optional inclusion, in the selection system, of a binding step involving protein L, A or any ligand, as well as stable non-antibody scaffolds and variants thereof.
  • the observed correlation between phage plaque size and V H expression yield means that one can utilize the present approach for acquiring high-expressing versions of proteins with otherwise poor or unsatisfactory expression from mutant phage display libraries. This application would be particularly appealing in the case of therapeutic proteins or expensive poor-expressing protein reagents where boosting protein expression would significantly offset protein production cost.
  • V L s and V H s are amenable to pentamerization and the pentamerization can be used to quickly convert a low affinity V L or V H monomer to a high affinity V L or V H pentamer.
  • Such pentamers are invaluable diagnostics and detection agents.
  • the binding of a V L or V H pentamer to its target can be detected by a reporter molecule such as an enzyme (for example, horse radish peroxidase or alkaline phosphatase), or a fluorescent molecule conjugated to the pentamer.
  • the binding of the pentamer can be detected by a secondary molecule which is conjugated to a reporter molecule.
  • the secondary molecule can be specific to the pentamer itself or to a tag thereof, such as a 6His tag or c-Myc tag.
  • a typical secondary molecule is an immunoglobulin.
  • V H s and protein A and V L s with protein L are fundamentally different from those between V H s and V L s with their target antigens.
  • the antigen binding of a V H or a V L involves three antigen binding loops which form the combining site of an antibody domain.
  • the protein A binding of a V H with protein A binding activity and a V L with protein L binding activity involve binding sites and residues on the antibody domains that are totally distinct from the antibody combining site.
  • a V H with protein A binding activity can simultaneously bind to protein A and its target antigen
  • a V L with protein L binding activity can simultaneously bind to protein L and its target antigen. Since the present V H s and V L s have affinity for protein A and L, respectively, protein A and L can be used as the secondary molecule for detection and diagnostic applications mentioned above.
  • the human V H and V L pentamers can also be used for therapy.
  • the protein A and L binding activity of the V H s and V L s can be used to detect bacteria which have protein A and/or L on their surfaces. Protein A is present on the surface of the pathogenic bacteria, Staphylococcus aureus. Thus, the V H s with protein A binding activity such as the ones described here can be used to detect S. aureus. Similarly, the V L monomers and V L P entamers with protein L binding activity can be used for the detection of bacteria, in particular pathogenic bacteria such as Peptostreptococcus magnus, which have protein L on their cell surface.
  • Protein L is implicated as a virulent factor in the pathogenesis of P. magnus (Ricci, S. et al., 2001) in humans. In vaginosis, protein L is thought to exert its effect by cross-linking surface associated IgE. V L monomers and/or pentamers with protein L binding activity have potential as therapeutics since they could interfere with the IgE cross-linking action of protein L.
  • Protein A is implicated as a virulent factor in the pathogenesis of S. aureus in humans (Fournier, B. et al., 2004). Its virulence has been attributed to its ability to interact with host components including binding to antibodies. V H monomers and/or pentamers with protein A binding activity have potential as therapeutics since they could interfere with the interaction of of protein A with host components.
  • plaque size was used as a means of identifying rare, naturally-occurring monomer V H s from the human V H repertoire ( Figure 1 ).
  • a phage library displaying human V H s with a size of 6 x 10 8 was constructed and propagated as plaques on agar plates. On the titer plates, the library consisted essentially of small plaques interspersed with some large ones.
  • V H -displaying phages PCR on twenty clones revealed that the small plaques corresponded to the V H -displaying phages while the large ones represented the wild type phages, i.e., phages lacking V H sequence inserts. None of the V H -displaying phages were found with large plaque morphology. This was not unexpected due to the paucity of the monomeric V H s in the human repertoire and the large size of the library. To facilitate the identification of monomeric V H s, it was decided to reduce the library to a manageable size and remove the interfering wild type phages with large-plaque-size morphology by panning the library against protein A which binds to a subset of human V H s from V H 3 family.
  • the most frequently-utilized germline V segment was DP-47 which occurred in over 50% of the V H s, but the most frequent clone (i.e., HVHP428; relative frequency 46%) utilized the V3-49 germline V segment.
  • HVHP429 with a DP-47 germline sequence was the second most abundant V H with a relative frequency of 21% ( Figure 2 ).
  • the V H CDR3 lengths ranged from 4 amino acids for HVHB82 to 16 amino acids for HVHP430 amino acids, with HVHP430 having a pair of Cys residues in CDR3.
  • HVHP44 While the positively-charged amino acid in HVHP44 was acquired by mutation, the one in HVHB82 was germline-encoded. Except for HVHP423 and HVHP44B, the remaining V H s had the germline residues at the key solubility positions: 37V/44G/45L/47W or 37F/44G/45L/47W (HVHP428); HVHP423 and HVHP44B had a V37F mutation. Mutations at other positions which are shown or hypothesized to be important in V H solubility included seven E6Q, three S35T/H, one R83G and one K83R, one A84P and one T84A and one M108L. Frequent mutations were also observed at positions 1 and 5 which included eleven E1Q, eight L5V/Q and one V5Q mutations.
  • V H s The aggregation tendency of the human V H s was assessed in terms of their oligomerization states by gel filtration chromatography and NMR (Table 2). All V H s were subjected to Superdex 75 gel filtration chromatography. Similar to a llama V H H, i.e., H11C7, all V H s gave a symmetric single peak at the elution volume expected for a monomer, and were substantially free of any aggregates (see the example for HVHP428 in Figure 3A . ln contrast, a typical human V H (i.e., BT32/A6) formed considerable amount of aggregates. For three of the V H s, a minor peak with a mobility expected for a V H dimer was also observed.
  • the state of protein aggregation was also assessed by use of an PFG-NMR diffusion experiment for the HVHP414 fragment and two isoforms, VH14 and VH14-cMyc- with and without the c-Myc sequence, of the HVHP414.
  • VH14 is a modified version of HVHP414 with a c-Myc N132E mutation and with an additional methionine residue at the N-terminus.
  • the PFG-NMR data (not shown) indicated that all the protein samples had expected monomeric molecular weights even at the relatively high protein concentrations used for NMR experiments.
  • V H s The stability of the V H s was further investigated in terms of their resistance to trypsin at 37°C integrity following long incubations at 37°C. Trypsin cleaves polypeptide amide backbones at the C-terminus of an Arg or a Lys residue. There are 9-13 Arg and Lys residues in the human V H s ( Figure 2 ). There is also an additional Lys residue in the C-terminal c-Myc tag which is susceptible to digestion by trypsin.
  • Figure 4a is an SDS-PAGE analysis of HVHP414 during trypsin digestion. Within 1 h the original band was completely converted to a single product which had a mobility expected for the V H with no c-Myc-HiS 5 tag.
  • V H s ranging in concentration from 0.32 mg/ml (HVHP428) to 3.2 mg/ml (HVHP420) were incubated at 37°C for 17 days. Their stability was subsequently determined in terms of oligomerization state and protein A binding. As shown by gel filtration chromatography, treatment of V H s at 37°C did not induce any aggregate formation: all V H s gave chromatogram profiles which were virtually identical to those of untreated V H s and stayed essentially as monomers (see the example for HVHP420; Figure 4c ) .
  • V H s namely, HVHP414 (1.2 mg/ml) and HVHP420 (3.2 mg/ml)
  • HVHP414 1.2 mg/ml
  • HVHP420 3.2 mg/ml
  • K D s of binding to protein A were determined by SPR (Data shown for HVHP420; Figure 4c inset) and compared to the K D s obtained for untreated V H s (Table 2).
  • the calculated K D s for the heat-treated V H s were 1.4 ⁇ M and 1.0 ⁇ M for HVHP414 and HVHP420, respectively. These values are essentially identical to the corresponding values for the untreated V H s (Table 2), demonstrating that 37°C treatment of V H s did not affect their native fold.
  • V H s may have been in a less compact, non-native fold during the 37°C-incubation periods and resumed their native fold upon returning to room temperature during gel filtration and SPR experiments is unlikely in light of the fact that the V H s were resistant to trypsin at 37°C (see above), a property typically associated for well folded native proteins.
  • the refolding efficiency ( RE ) of the human V H s was investigated by comparing the K D s of the binding of the native (K D n) and heat-treated, refolded (K D ref ) V H s to protein A (Tanha, J. et al., 2002) .
  • K D n native
  • K D ref heat-treated, refolded
  • V H s protein A
  • the ratio of K D n to K D ref gives a measure of V H RE.
  • Figure 5 compares sensorgrams for HVHP423 binding to immobilized protein A in native (thick lines) and refolded (thin lines) states at several selected V H concentrations. As can be seen, binding of the refolded V H to protein A is less in all instances, indicating that the unfolding is not fully reversible. For each of the 14 V H s, protein A binding in both native and refolded states was measured at several concentrations, and the K D s and subsequently RE s were determined ( Table 2; K D ref values are not shown). The K D s and RE s of two anti-idiotypic llama V H Hs, H11 F9 and H11 E32, which were used as references, were also determined.
  • V H s had RE s in the range of 92%-95%, similar to the RE s for H11F9 and H11B2, 95% and 100%, respectively. Another five had RE s in the range of 84%-88% and three over 70%. Only two had significantly lower RE: HVHP413 (52%) and HVHP421 (14%).
  • V H Hs examined previously had RE around 50% van der Linden, R. H. et al ., 1999
  • V H phage display library construction and panning Human V H phage display library construction and panning.
  • cDNA was synthesized from human spleen mRNA (Ambion Inc., Austin, TX) using random hexanucletide primers and First Strand cDNATM kit (GE Healthcare, Baie d'Urfé, QC, Canada).
  • V H genes with flanking C H sequences were amplified by polymerase chain reaction (PCR) in nine separate reactions using V H framework region 1 (FR1 )-specific primers and an immunoglobin M-specific primer (de Haard, H. J . et al., 1999) .
  • V H genes were get-purified and used as the template in the second round of PCR to construct V H genes using the FR1- and FR4-specific primers (de Haard, H. J. et al., 1999) that also introduced flanking Apal I and Not I restriction sites for cloning purposes.
  • the resultant V H repertoire DNAs were cloned into fd-tetGlllD phage vector and a V H phage display library was constructed (Tanha, J. et al., 2001) . Panning against protein A (Amersham Biosciences Inc.) was performed as described (Tanha, J. et al . , 2001) .
  • V H Hs H11C7, H11 F9 and H11 B2 were isolated from a llama V H H phage display library by panning against H11 scFv as described (Tanha, J. e t al ., 2002) .
  • V H expression and purification V H s were cloned into pSJF2 expression vectors by standard cloning techniques (Sambrook, J. Fritsch E. F. and Maniatis T, 1989) . Periplasmic expression of sdAbs and subsequent purification by immobilized metal affinity chromatography (IMAC) were performed as described ( Muruganandam , A. et al., 2002) . Protein concentrations were determined by A 280 measurements using molar absorption coefficients calculated for each protein (Pace, C. N. et al., 1995). Gel filtration chromatography of the purified V H s was performed on a Superdex 75 column (GE Healthcare) as described (Deng, S. J. et al., 1995) .
  • IMAC immobilized metal affinity chromatography
  • Immobilizations were carried out at concentrations of 25 ⁇ g/ml (protein A) or 50 ⁇ g/ml (Fab) in 10 mM sodium acetate buffer pH 4.5, using the amine coupling kit provided by the manufacturer.
  • 4100 RUs of 50 ⁇ g/ml H11 scFv or 3000 RUs of 10 ⁇ g/ml Se155-4 IgG reference were immobilized as described above.
  • V H s or V H Hs were denatured by incubation at 85°C for 20 min at 10 ⁇ g/ml concentrations. The protein samples were then cooled down to room temperature for 30 min to refold and were subsequently centrifuged in a microfuge at 14,000 rpm for 5 min at room temperature to remove any protein precipitates.
  • K D n is the K D of the native protein and K D ref is the K D of the refolded protein.
  • Tryptic digest experiments 3 ⁇ l of a freshly prepared 0.1 ⁇ g/ ⁇ l sequencing grade trypsin (Hoffmann-La Roche Ltd., Mississauga, ON, Canada) in 1 mM HCI was added to 60 ⁇ g V H in 100 mM Tris-HCI buffer pH 7.8. Digestion reactions were carried out in a total volume of 60 ⁇ l for 1 h at 37°C and stopped by adding 5 ⁇ l of 0.1 ⁇ g/ ⁇ l trypsin inhibitor (Sigma, Oakville, ON, Canada).
  • sdAbs Single-domain antibodies
  • PBS buffer for 17 days.
  • the protein samples were spun down in a microfuge at maximum speed for 5 min even in the absence of any visible aggregate formation.
  • the samples were then applied onto a Superdex 75 size exclusion column (GE Healthcare) and the monomeric peaks were collected for SPR analysis against protein A.
  • SPR analyses were performed as described above except that 500 RUs of protein A or reference Fab was immobilized and that immobilizations were carried out at concentration of 50 ⁇ g/ml.
  • NMR experiments - V H samples for NMR analysis were dissolved in 10 mM sodium phosphate, 150 mM NaCI, 0.5 mM EDTA, and 0.02% NaN 3 at pH 7.0. The protein concentrations were 40 ⁇ M - 1.0 mM. All NMR experiments were carried out at 298 K on a Bruker Avance-800 or a Bruker Avance-500 NMR spectrometer. One-dimensional (1D) 1 H NMR spectra were recorded with 16,384 data points and the spectral widths were 8,992.81 Hz at 500 MHz and 17,605.63 Hz at 800 MHz, respectively.
  • V L phage display library construction and panning Human V L phage display library construction and panning.
  • cDNAs were synthesized from human spleen mRNA as described above for the human V H s. The cDNA was used as template in PCR to amplify V L genes in 50 ⁇ l reaction volumes using six V ⁇ back primers, 11 V ⁇ back primers (de Haard, H. J. et al., 1999). four V ⁇ For primers and two V ⁇ For primers (Sblattero, D. et al., 1998). The back and forward primers were modified to have flanking Apa Ll and Not l restriction sites, respectively, for subsequent cloning purposes. Forward primers were pooled together in ratios which reflected their degree of degeneracy.
  • V ⁇ genes were PCRed in 11 separate reactions using the pooled V ⁇ For primers and 11 individual V ⁇ back primers. Similarly, V ⁇ genes were amplified in 6 separate reactions using the pooled V ⁇ For primers and 6 individual V ⁇ back primers.
  • the PCR products were pooled, gel purified and digested with Apa LI and Not I restriction endonucleases.
  • the library was constructed as described for human V H s. Plaque PCR was performed on individual library colonies and the amplified V L genes were sequenced as described (Tanha, J. et al., 2003). Panning against protein L (Biolynx Inc., Brockville, ON, Canada) and germline sequence assignment of the selected V L s were performed as described above for human V H library.
  • V L expression and purification V L expression and purification.
  • V L expression, purification, concentration determination and gel filtration chromatography were carried out as described for V H s in "V H expression and purification,".
  • V L and V H pentamers Expression and purification of V L and V H pentamers. Specific primers were used in a standard PCR to amplify HVHP328 V H and HVLP335 V L genes. Standard cloning techniques were used to clone the HVHP328 and HVLP335 genes in fusion with VT1B pentamerization domain gene in an expression vector to yield HVHP328PVT2 and HVLP335PTV2 pentamers, (Zhang, J. et al., 2004). Pentamers were expressed and purified as described (Zhang, J. et al., 2004). Protein concentrations were determined as above.
  • V L s Surface plasmon resonance of V L s.
  • the binding kinetics for the interaction of the V L s to protein L were determined by SPR using BIACORE 3000 biosensor system (Biacore, inc., Piscataway, NJ). 680 RUs of protein L or 870 RUs of a Fab reference were immobilized on research grade CM5 sensor chips (Biacore). Immobilizations were carried out at a protein concentration of 50 ⁇ g/ml in 10 mM acetate buffer pH 4.5 using the amine coupling kit supplied by the manufacturer.
  • a single S. aureus colony from a BHI plate was used to inoculate 15 mL of BHI media.
  • the bacteria were grown overnight at 37°C at 200 rpm.
  • the culture was spun down in a swinging bucket, Sorvall RT6000B refrigerated centrifuge at 4000 rpm for 10 min, the supernatant was removed and the cell pellet was re-suspended in PBS buffer.
  • the cells were re-spun, the supernatant was removed and the cell pellet was re-suspended again in PBS buffer.
  • the cells were diluted to an A 600 of 1.0, and serial dilutions of the cells were spread on BHl plates at 37°C for overnight growth.
  • Well 12 (blank) had only PBS. The total volume in each well was 50 ⁇ l. Subsequently, 1 x 10 8 S. aureus cells in 50 ⁇ lPBS was added to all wells and the plate was incubated overnight at 4°C. To have a permanent record of the results, a picture was taken from the plate in the morning. For the pentamer control experiment, HVHP328PVT2 was replaced with the V L pentamer, HVLP335PTV2. In the cell control experiments, the same two sets of experiments were repeated with E. coli TG1 cells.
  • HVLP325, HVLP335 and HVLP351 occurred at frequency of 3, 4 and 2, respectively. Except for HVLP389 which is of lambda class (subgroup V ⁇ 1, germline 1b), the remaining 31 V L s belonged to the V ⁇ class. Of the 31 kappa V L s, 24 fall within the V ⁇ lll subgroup and 7 within the V ⁇ 1 subgroup. Sixteen of the 24 V ⁇ lll sequences utilize L6 germline sequence with the remaining utilizing A27, L2 and L6 germline sequences. The V ⁇ 1 subgroup V L s are originated from 02/012 or A30 germline sequence. Noticeable mutations occurred at position 96.
  • the germline amino acids at this position are aromatic and hydrophobic amino acids Trp, Phe, Tyr, Leu or Ile for kappa V L s and Tyr, Val or Ala for lambda V L s. But in the selected pool of kappa V L s only 5 out of 31 have their germline amino acids at position 96: HVLP325, HVLP349, HVLP388, HVLP3109 and HVLP393. 21 amino acids at position 96 are charged of which 20 are positively-charged: Arg, Lys or His. Two amino acids are Pro, one Gin, one Ser and one Thr.
  • V L s Of seven kappa V L s analyzed by gel filtration chromatography for monomericity, six which had Arg or Lys at position 96 were also monomers, whereas HVLP325 with the germline amino acid Leu at position 96 formed aggregates (see below). Similarly, HVLP389 which was of the lambda class and had a germline mutation to Ser was also monomeric (see below). These data correlates the deviation from the germline amino acids at position 96 (27 out of 32) with improved biophysical properties of V L s such as monomericity.
  • V L s of the kappa class had their last three residues (105-107) replaced with amino acids Thr, Val and Leu which are only found in lambda V L s. These substitutions may have had a role in improving the biophysical properties of the kappa V L s, resulting in the selection of the aforementioned V L s over the parental clones with the original kappa residues at position 105-107.
  • HVLP324, HVLP325, HVLP335, HVLP342, HVLP351, HVLP364, HVLP389 and HVLP3103 (Table 6). All were expressed in good yields ranging from 6.2 mg for HVLP324 to around 75 mg for HVLP335 and HVLP364.
  • V L s The aggregation tendency of the human V L s was assessed in terms of their oligomerization state by gel filtration chromatography.
  • V L s were subjected to Superdex 75 gel filtration chromatography at a concentration of 0.6 mg/ml. All except HVLP325 were essentially free of aggregates and gave symmetric single peaks with the mean apparent molecular mass of 12.7 kDa (range, 6.2-19.2 kDa) ( Figure 7A and Table 3 ). This is in agreement with the expected molecular mass for monomeric V L s, 13.4-13.8 kDa. Variation in apparent molecular mass for single-domain antibodies has been reported previously (Jespers, L. et al., 2004a; (Stevens, F. J.
  • HVLP325 the aggregates formed 11% of the total protein (aggregate plus monomer).
  • HVLP351, HVLP342, HVLP335 and HVLP3103, were still monomer when tested at their highest concentration available, i.e., 0.89 mg/ml 1.0 mg/ml, 4.9 mg/ml and 5.9 mg/ml, respectively ( Figure 7B )
  • V L s were subjected to Superdex-75 chromatography prior to BIACORE analysis and purified monomer peaks collected even in the absence of any evidence of aggregated material.
  • SPR analysis all selected V L s bound to protein L ( Figure 8 ). This was not unexpected since the V L s were isolated by panning against protein L. For all, the K D s of binding to protein L were in 0.6-3 ⁇ M (Table 3). HVLP324 and HVLP342 had additional smaller K D s, 10 nM and 40 nM, respectively. Low affinity and high affinity bindings of V L s of V ⁇ l subgroup to protein L have been reported previously (Reference). Both, HVLP324 and HVLP342, belong to V ⁇ l subgroup (Table 3). As expected, the kinetic and equilibrium data were consistent with the monomeric peak being indeed monomeric.
  • HVHP328PVT2 pentamer to protein A and HVLP335PTV2 pentamer to protein L were determined by surface plasmon resonance ( Figure 9 ).
  • the association rates were independently calculated from plots of k obs versus concentration. More than one dissociation rate (k d ) could be calculated due to the heterogeneity in multivalent binding amongst the pentamer population. Therefore, more than one equilibrium dissociation constant, K D , could be obtained.
  • HVHP328PTV2 and HVLP335PTV2 had minimum K D s of 2 nM and 200 pM, respectively (Table 4). With slower k d s, HVHP328PTV2 and HVLP335PTV2 had K D s as low as 900 and 90 pM, respectively.
  • the protein A and L binding activity of the V H s and V L s can be used to detect bacteria which have protein A and/or L on their surfaces. This is possible if the V H s and V L s are soluble and monomeric (lack of tendency to aggregate) such as the V HS and V L s here. Variable domains derived from antibodies which lack light chains such as camelid heavy chain antibodies or nurse shark and wobbegong shark lgNARs are naturally soluble and monomeric. From these, those with protein A and L binding activity.can also be used to detect bacteria which have protein A and/or L on their surfaces. Protein A is present on the surface of the pathogenic bacteria, Staphylococcus aureus.
  • the V H S with protein A binding activity such as the ones described here can be used to detect S. aureus.
  • a constant number of bacterial cells were incubated with two-fold dilutions of HVHP328PVT2 in microtiter wells (wells 1-11) ( Figure 10 ).
  • Well 12 had buffer instead of the pentamer. lf the V H S bind to the bacterial cells, then the pentamer because of its multimeric nature should be able to cross-link the cells and results in cell agglutination.
  • the agglutinated cells will appear as diffused cells in a microtiter well ( Figure 10 ). ln the absence of any binding, no agglutination should occur, hence no agglutination, and the cells will appear as a dot at the bottom of the well.
  • the pentamer binds to the S. aureus, since there is agglutination of cells. The agglutination is observed up to well 7. Beyond well 7 the concentration of the pentamer is too low for binding, hence no agglutination.
  • the control V L pentamer does not show any agglutination, demonstrating the specificity of the V H pentamer to S. aureus ( Figure 10 ).
  • V H pentamer as expected does not agglutinate E. coli (TG1 strain) or Salmonella cells (data not shown).
  • V L monomers and V L pentamers with protein L binding activity can be used for the detection of bacteria, in particular pathogenic bacteria such as Peptostreptococcus magnus, which have protein L on their cells surface.
  • a method of identifying target polypeptides comprising:
  • target polypeptides are human V H or V L antibody fragments.
  • target polypeptides are selected from the group consisting of: single-chain T-cell receptors, T-cell receptor domains, transferin, lipocalins, kunitz domains, ankyrin repeats, and cytotoxic T-lymphocyte-associated antigen.
  • target polypeptides are proteinaceous diagnostic and biochemical reagents.
  • a nucleic acid sequence that encodes a polypeptide as described in embodiment 24 or 25 is provided.
  • a polypeptide comprising the CDR3 portion of an amino acid sequence as described in embodiment 24.
  • a nucleic acid sequence that encodes a polypeptide as described in embodiment 27 is provided.
  • a polypeptide comprising the FR1 1 portion of an amino acid sequence as described in embodiment 24.
  • a nucleic acid sequence that encodes a polypeptide as described in embodiment 29 is provided.
  • a polypeptide comprising the FR2 portion of an amino acid sequence as described in embodiment 24.
  • a nucleic acid sequence that encodes a polypeptide as described in embodiment 31 is provided.
  • a polypeptide comprising the FR3 portion of an amino acid sequence as described in embodiment 24.
  • a nucleic acid sequence that encodes a polypeptide as described in embodiment 33 is provided.
  • a polypeptide comprising the FR4 portion of an amino acid sequence as described in embodiment 24.
  • a nucleic acid sequence that encodes a polypeptide as described in embodiment 35 is provided.
  • a polypeptide comprising the CDR2 portion of an amino acid sequence as described in embodiment 24.
  • a nucleic acid sequence that encodes a polypeptide as described in embodiment 37 is provided.
  • a polypeptide comprising the CDR1 portion of an amino acid sequence as described in embodiment 24.
  • a nucleic acid sequence that encodes a polypeptide as described in embodiment 39 is provided.
  • a V H antibody fragment comprising at least one amino acid sequence selected from the group consisting of: SEQ lD NO:8-22.
  • a V L antibody fragment comprising at least one amino acid sequence selected from the group consisting of: SEQ lD NO:23-54.
  • a V H antibody fragment comprising at least two amino acid sequences as described in embodiment 41.
  • a V L antibody fragment comprising at least two amino acid sequences as described in embodiment 42.
  • a V H antibody fragment comprising at least three amino acid sequences as described in embodiment 41.
  • a V L antibody fragment comprising at least three amino acid sequences as described in embodiment 42.
  • a multimer comprising at least two V H antibody fragments as described in embodiment 41, 44 or 47.
  • a multimer comprising at least two V L antibody fragments as described in embodiments 42, 45 or 48.
  • a multimer comprising at least one V H antibody fragment as described in embodiments 41, 44 or 47, and at least one V L antibody fragment as described in embodiments 42, 45 or 48.
  • a dimer comprising two V H antibody fragments as described in embodiment 41, 44 or 47.
  • a dimer comprising two V L antibody fragments as described in embodiment 42, 45 or 48.
  • a trimer comprising three V H antibody fragments as described in embodiment 41, 44 or 47.
  • a trimer comprising three V L antibody fragments as described in embodiments 42, 45 or 48.
  • a pentamer comprising five V H antibody fragments as described in embodiments 41, 44 or 47.
  • a pentamer comprising five V L antibody fragments as described in embodiment 42, 45 or 48.
  • V H antibody fragment identified by the method described in embodiment 1.
  • V H antibody fragment as described in embodiment 59 that is of human origin.
  • V H antibody fragment as described in embodiment 59 having an amino acid sequence that deviates from the corresponding parental germline sequence as shown in Table 1.
  • V H antibody fragment as described in embodiment 59 belonging to the V H 3 family is A V H antibody fragment as described in embodiment 59 belonging to the V H 3 family.
  • V H antibody fragment as described in embodiment 59 and having a residue other than valine at position 37 of its amino acid sequence.
  • V H antibody fragment as described in embodiment 63 having a phenylalanine residue at position 37 of its amino acid sequence.
  • V H antibody fragment as described in embodiment 63 having a tyrosine residue at position 37 of its amino acid sequence.
  • a V H antibody fragment as described in embodiment 59 having a mutation at a position selected from the group consisting of positions 1,5,6,35,83,84,84a and 108 of its amino acid sequence.
  • V H antibody fragment as described in embodiment 59 which binds to protein A.
  • V L antibody fragment identified by the described in embodiment 1.
  • a V L antibody fragment as described in embodiment 70 belonging to the kappa class is also included in embodiment 70.
  • a V L antibody fragment as described in embodiment 70 having an arginine, proline, lysine, threonine, leucine, serine, tyrosine, glutamic acid, glutamine or histidine residue at position 96 of its amino acid sequence.
  • V L antibody fragment as described in embodiment 70 which binds to protein L.
  • a nucleic acid sequence that encodes the polypeptide sequence described in embodiment 80 is A nucleic acid sequence that encodes the polypeptide sequence described in embodiment 80.
  • a display library constructed using polypeptide sequences as described in embodiments 24, 59 or 70.
  • a display library constructed using V H or V L antibody fragments as described in embodiments 41, 42, 44, 45, 47 or 48.
  • a display library as described in embodiment 82, 83, 84, 85 or 86 that is a ribosome display, ARM ribosome display, yeast display, bacterial cell display or in vitro compartmentalization library.
  • a method for producing polypeptides with desirable biophysical properties comprising the steps of a) providing at least one first nucleic acid sequence that encodes an antibody fragment as described in embodiment 41,42,44,45,47,48, 59 or 70 or that encodes a polypeptide sequence described in embodiment 24,27,37 or 39, and having a first desirable property; b) providing at least one second nucleic acid sequence that encodes an antibody fragment having a second desirable property; c) cutting the at least one first and at least one second nucleic acid sequences into random fragments; d) reassembling the random fragments; e) expressing the random fragments; and f) screening the expressed random fragments for the first and second desirable properties.
  • a method as described in embodiment 89, wherein the second desirable property is selected from the group consisting of solubility, stability, monomericity, high expression, human origin and binding specificity.
  • a method as described in embodiment 89, where screening of the reassembled random fragments is accomplished by ribosome display, yeast display, phage display, bacterial cell display, ARM ribosome display or in vitro compartmentalization.
  • a pharmaceutical composition comprising the antibody fragment described in embodiment 59 or 70, and a pharmaceutically suitable agent.
  • a pharmaceutical composition comprising the polypeptide sequence described in embodiment 24 or 80, and a pharmaceutically suitable agent.
  • a recombinant vector comprising the nucleic acid sequence described in embodiment 26 or 81.

Abstract

Polypeptides with desirable biophysical properties such as solubility, stability, high expression, monomericity, binding specificity or non-aggregation, including monomeric human VHs and VLS, are identified using a high throughput method for screening polypeptides, comprising the steps of obtaining a phage display library, allowing infection of a bacterial lawn by the library phage, and identifying phage which form larger than average plaques on the bacterial lawn. Sequences of monomeric human VHS or VLS are identified, which may be useful for immunotherapy or as diagnostic agents. Multimer complexes of human VHS and VLS are also identified. The VHS and VLS identified many be used to create further libraries for identifying additional polypeptides. Further, the VHS and VLS may be subjected to DNA shuffling to select for improved biophysical properties.

Description

    Field of the Invention
  • This invention relates to the isolation, identification and manipulation of polypeptides, especially monomeric human antibody fragments.
  • Background of the Invention
  • Antibodies in vertebrates are typically composed of paired heavy (H) and light (L) chains. The first domain of the combined H and L chains, the VH and VL, are more variable in sequence, and this is the portion of the antibody that recognizes and binds to the antigen. The VH and VL domains recognize the antigen as a pair.
  • The immune repertoire of camelidae (camels, dromedaries and llamas) is unique in that it possesses unusual types of antibodies referred to as heavy-chain antibodies (Hamers, Casterman C. et al., 1993). These antibodies lack light chains and thus their combining sites consist of one domain, termed VHH.
  • Recombinant VHH single-domain antibodies (sdAbs) provide several advantages over single-chain Fv (scFv) fragments derived from conventional four-chain antibodies. While sdAbs are comparable to their scFv counterparts in terms of affinity, they outperform scFvs in terms of solubility, stability, resistance to aggregation, refoldability, expression yield, and ease of DNA manipulation, library construction and 3-D structural determinations. Many of the aforementioned properties of VHH sdAbs are desired in applications involving antibodies.
  • However, the non-human nature of VHHs limits their use in human immunotherapy due to immunogenicity. In this respect, human VH and VL sdAbs are ideal candidates for immunotherapy applications because they are expected to be least immunogenic.
  • Human VHs and VLs, however, are by and large prone to aggregation, a characteristic common to VHs and VLs derived from conventional antibodies (Davies, J. et al., 1994;Tanha, J. et al., 2001;Ward, E. S. et al., 1989). Thus, attempts have been made to obtain monomer human VHs and VLs suitable for antibody applications. Such VHs and VLs have also displayed other useful properties typical of VHHs such as high expression yield, high refoldability and resistance to aggregation. Synthetic libraries built on these VHs and VLs as library scaffolds might serve as a promising source of therapeutic proteins.
  • Camelization as well as Ilamination which involves incorporating key solubility residues from camel and llama VHHs, respectively, into human VHs or VLs have been employed to generate monomeric human VHs and VLs. Synthetic sdAb libraries constructed based on these VHs and VLs and generated by CDR randomization were shown to be functional in terms of yielding binders to various antigens (Davies, J. et al., 1995;Tanha, J. et al., 2001).
  • In another approach, fully human monomeric VHs and VLs were isolated from human synthetic VH and VL libraries without resorting to engineering of the sort mentioned above. In one experiment a monomeric human VH, was discovered when a human VH library was panned against hen egg lysozyme (Jespers, L. et al., 2004b). More recently, a selection method based on reversible unfolding and affinity criteria yielded many monomeric VHs from synthetic human VH libraries (Jespers, L. et al., 2004a). This finding underlined the fact that an appropriate selection method is key to efficient capturing of rare monomer human VHs with desirable biophysical properties.
  • Objects of the Invention
  • A first object of the invention is to provide a high throughput screening method for identifying polypeptides, especially antibody fragments, with improved biophysical properties, including solubility, high expression, and/or stability (such as high refolding after thermal denaturation, high resistance to chemical denaturant, and high resistance to proteases, in particular gastrointestinal proteases such as trypsin).
  • A second object of the invention is to provide a high throughput screening method for identifying monomeric human VHs and VLs.
  • A third object of the invention is to identify, isolate and characterize monomeric human VHs and VLs.
  • A fourth object of the invention is to construct and characterize multimers of antibody fragments, especially monomeric human VHs and VLs.
  • A fifth object of the invention is to construct display libraries from polypeptides, especially antibody fragments, and most especially monomeric human VHs and VLs.
  • A sixth object of the invention is to provide a DNA shuffling method for producing polypeptides, especially antibody fragments, and most especially monomeric human VHs and VLs with improved biophysical properties.
  • Summary of the Invention
  • A method is provided for isolating polypeptides, preferably antibody fragments, and most preferably human VHs and VLs with desirable biophysical properties (solubility, stability, high expression, monomericity, non-aggregation, binding specificity). The method includes the steps of obtaining a phage display library capable of expressing a variety of polypeptide sequences, allowing infection of a bacterial lawn by the library phage, and identifying phage which form larger than average plaques on the bacterial lawn. The phage are then isolated, and steps are taken to sequence or otherwise characterize the polypeptide sequences.
  • The invention also provides for polypeptides, especially monomeric human VHs and VLs, identified by the above method, which may be useful for immunotherapy, and/or as diagnostic or detection agents. The monomeric human VHs and VLs may also be combined to form dimers, trimers, pentamers or other multimers, which may be useful for immunotherapy and/or as diagnostic or detection agents.
  • The polypeptides identified by the above method, including human VHs and VLs, can be manipulated by methods such as DNA shuffling to select for improved biophysical properties such as solubility, stability, monomericity, high expressibility, binding specificity and human origin.
  • The polypeptides identified by the above method, including human VHs and VLs, may also be used to generate further display libraries, which can then in turn be used to isolate further polypeptides by the above method.
  • In a first aspect, the present invention provides a method of identifying target polypeptides , comprising a) obtaining a phage display library capable of expressing a variety of polypeptide sequences, b) allowing infection of a bacterial lawn by the library phage and c) identifying phage which form larger than average plaques on the bacterial lawn.
  • In a second aspect, the present invention provides polypeptide having an amino acid sequence selected from the group consisting of: SEQ ID NO:8-54
  • In a third aspect, the present invention provides a VH antibody fragment comprising at least one amino acid sequence selected from the group consisting of: SEQ ID NO:8-22.
  • In a fourth aspect, the present invention provides a VL antibody fragment comprising at least one amino acid sequence selected from the group consisting of: SEQ ID NO:23-54.
  • In a fifth aspect, the present invention provides A method for producing polypeptides with desirable biophysical properties, comprising the steps of a) providing at least one first nucleic acid sequence that encodes an antibody fragment as claimed in claim 41,42,44,45,47,48, 59 or 70 or that encodes a polypeptide sequence as claimed in claim 24,27,37 or 39, and having a first desirable property; b) providing at least one second nucleic acid sequence that encodes an antibody fragment having a second desirable property; c) cutting the at least one first and at least one second nucleic acid sequences into random fragments; d) reassembling the random fragments; e) expressing the random fragments; and f) screening the expressed random fragments for the first and second desirable properties.
  • Detailed Description of the Drawings Figure legends
    • Figure 1 . A pictorial representation of selected example results: The contrast in plaque size between phages displaying a soluble VH (HVHP428) and those displaying an insoluble one (BT32/A6). The photo depicts a part of the bacterial lawn agar plate which was magnified to enhance plaque visualization. Although the plate contained an equal number of each of the two plaque types, the photo essentially contains the large, HVHP428 plaques. The majority of the BT32/A6 plaques were too small to produce clear, well-defined images in the photo. The plaques marked by arrows, thus, represent a minor proportion of BT32/A6 phages which were large enough to be visible in this image. Asterisks marks representative plaque sizes for HVHP428 phages. The identities of plaques were determined by DNA sequencing.
    • Figure 2 . Amino acid sequence of the human VHs selected based on affinity for protein A and plaque size. The dots in the sequence entries indicate amino acid identity with HVHP2M10 or HVHP44. Dashes are included for sequence alignment. Residues at the key solubility positions and residue 57T which associates with VHs/VHHs with protein A binding property are in bold. The Kabat numbering system is used. The total "frequency" value is 114. CDR = complementarity determining region; FR = framework region; gln seq = germline sequence
    • Figure 3 . Aggregation tendencies of the human VHs. Gel filtration chromatograms comparing the oligomerization state of a human VH isolated in this study (HVHP428) to that of a llama VHH (H11C7) and a typical human VH (BT32/A6). The peak eluting last in each chromatogram corresponds to monomeric VH. The dimeric H11C7 peak is marked by an arrow. B, One-dimensional 1H NMR spectra of HVHP414 at 800 MHz (i), HVHP423 at 500 MHz (ii) and HVHP428 at 800 MHz (iii). The spectra in the left panel are scaled up by a factor of two to enable better viewing of low-intensity signals.
    • Figure 4 . Stability of the human VHs in terms of their resistance to trypsin at 37°C and integrity following long incubation at 37°C. A, SDS-PAGE comparing the mobilities of the untreated and trypsin-treated HVHP414 VH at 15, 30 and 60 min relative to a 21 kDa marker. HVHP414-cMyc denotes HVHP414 VH lacking the c-Myc. B, Molecular mass profiles obtained by mass spectrometry of untreated and trypsin-treated (60 min) HVHP414 VH. The mass spectrometry profile of the treated VH is superimposed onto that for the untreated one to provide a better visual comparison. The experimental molecular mass of the untreated VH is 14,967.6 Da, which is essentially identical to the expected molecular mass, 14,967.7 Da. The observed molecular mass of the trypsin-treated VH (13,368.5 Da) indicates loss of 13 amino acids at the C-terminus by cleavage at K (Lys) in the c-Myc tag to give an expected molecular mass of 13,368.0 Da. The trypsin cleavage site is shown by a vertical arrow above the amino acids sequence of HVHP414. C, Gel filtration chromatograms comparing the oligomerization state of the 37°C-treated HVHP420 VH (upper profile) to that of untreated VH (lower profile). The chromatograms were shifted vertically because they were indistinguishable when superimposed. The major and minor peaks in each chromatogram correspond to monomeric and dimeric VHs, respectively. The dimeric VH constitutes 3% of the total protein. The inset shows the sensorgram overlays for the binding of 37°C-treated HVHP420 to protein A at various concentrations. The VHs used for temperature stability studies were from stocks which had already been at 4°C for several months.
    • Figure 5 . Sensogram overlays showing the binding of native (thick lines) and refolded (thin lines) HVHP423 to immobilized protein A at 75, 100, 150 and 200 nM concentrations. KDn and KDref were calculated from respective sensograms and used to determine RE as described below.
    • Figure 6 . Amino acid sequences of the human VLs selected based on affinity for protein L and plaque size. The dots in the sequence entries indicate amino acid identity with HVLP333. Dashes are included for sequence alignment. See the V BASE (http://vbase.mrc-cpe.cam.ac.uk/index.php?module=pagemaster&PAGE_user_op=view_page& PAGE_id=7&MMN_position=5:5) for sequence numbering and CDR designation. L6, A27, L2, L16, 02/O12, A30 and 1b are V germline designation. J germline designations are in the brackets. NF, not found.
    • Figure 7 . Size exclusion chromatograms of human VL domains. In A, the VLs were applied at a concentration of 0.6 mg/ml. In B, the VLs were applied at their highest concentration available: HVLP342, 1.0 mg/ml; HVLP3103, 5.9 mg/ml; HVLP335, 4.9 mg/ml; HVLP351, 0.89 mg/ml. "#"and "*" represent aggregate and monomer peaks, respectively. The aggregates elute in the exclusion volume. The peak marked by an arrow in the HVLP342 panel (B) is the carry over from a previous run.
    • Figure 8 . Sensorgram overlays showing the binding of VLs to immobilized protein L at concentrations of 0.2, 0.5, 0.75, 1, 2, 3, 5 and 10 µM (HVLP389, HVLP351 and HVLP364); 1, 2, 3, 5, 7.5 and 10 nM (HVLP342); 0.2, 0.5, 1, 2, 3, 5 and 10 µM (HVLP335); 0.2, 0.5, 1, 1.5, 2 and 5 µM (HVLP325), 0.2, 0.5, 0.75, 1, 1.5, 2, 3 and 5 µM (HVLP3103) and 1, 2, 3, 4, 5 and 6 nM (HVLP324). The sensorgrams for HVLP324 and HVLP342 bindings to the low affinity site of protein L are not included but the calculated KDs are recorded in Table 3.
    • Figure 9 . Bindings of HVHP328PTV2 to protein A and HVLP335PTV2 to protein L in surface plasmon resonance experiments. (A) Sensorgram overlays showing the binding of HVH28PTV2 to immobilized protein A at 1, 2, 3, 4, 6, 8 and 10 nM concentrations. (B) Sensorgram overlays showing the binding of HVLP335PTV2 to immobilized protein L at 1, 2, 2.5, 3, 3.5, 4 and 4.5 nM concentrations. The binding data are recorded in Table 4.
    • Figure 10 . Figure showing the results of the microagglutination experiments with S. aureus cells. The concentration of the pentamers decreases two-fold from well 1 to well 11 with well 12 having the pentamers replaced with PBS buffer. The top row wells contain HVHP328PTV2 pentamer and the bottom ones HVLP335PTV2 pentamer. The concentrations of the pentamers in wells 1 to 6 are 215, 108, 54, 27, 13 and 7 µg/ml, respectively.
    Detailed Description of the Invention
  • It is desirable to identify polypeptides, especially antibody fragments, that are of human origin, soluble, stable, resistant to aggregation, refoldable, highly expressed, easily manipulated at the DNA level, ideal for library construction and for 3-D structural determinations. Such antibody fragments are useful for a wide variety of immunotherapeutical applications, and also as diagnostic and detection agents. Human monomeric VH and VL antibodies are of particular interest, as they are likely to have many of the above-mentioned properties.
  • Polypeptides with the above-mentioned properties may be identified by high throughput screening of libraries capable of expressing a variety of polypeptide sequences. For example, phage display libraries (preferably filamentous phage such as M13 or fd) may be screened by infecting a field of bacteria susceptible to the phage (a bacterial lawn) with the phage, then determining which phages have successfully lysed the bacteria by looking for clear, bacteria-free areas known as plaques. Phages displaying monomeric Ilaminated VHs and VLs form larger plaques on bacterial lawns than phages displaying fully human VHs with aggregation tendencies. Thus, plaque size may be used as a means of identifying rare, naturally-occurring monomer VHs and VLsfrom the human VH repertoire.
  • The method disclosed herein is also useful in identifying soluble, stable (stability covers a number of characteristics, including but not limited to high thermal refolding efficiency, high melting temperature, maintaining functionality after long (several days) incubation at 37°C, resistant to chemical denaturants, resistant to proteases, having a long shelf life at below 0°C, and 4°C, and at room temperature, maintaining functionality in intracellular environments, and maintaining functionality inside the human body, such as in the bloodstream) and high expressing proteins of differing origins, including:
    1. 1. VHs, VLs, Fabs, scFvs and whole antibodies such as IgGs, more specifically human ones
    2. 2. Protein variants based on non-antibody scaffolds single-chain T-cell receptors, T-cell receptor domains, transferin, lipocalins, kunitz domains, ankyrin repeats, and cytotoxic T-lymphocyte-associated antigen (CTLA-4), including human ones
    3. 3. Vaccines such as viral and bacterial protein vaccines
    4. 4. Therapeutic proteins, e.g., insulin, growth hormone, arythropoietin
    5. 5. Proteinacious diagnostic and biochemical reagents, e.g., protein A, protein G.
  • Once polypeptides have been identified by this method, they can be used to construct additional libraries. This is done by selecting a nucleic acid sequence of, for example, a VH. Oligonucleotides with randomized codons are created and incorporated into the VH sequence. Thus, each unique oligonucleotide is incorporated into a VH gene, and the modified VH genes constitute a library of sequences with slight variations. Typically, the oligonucleotides are designed such that the CDRs or loops of the VH are randomized. For example, one, two or all three of VH CDRs may be randomized. The VH library is then cloned into an appropriate vector, depending on the type of library to be used, and the nucleic acid sequences are expressed as polypeptides. The library is screened for molecules that bind to the library polypeptides, typically by panning. The libraries may be phage display libraries, or other display libraries such as ribosome display and yeast display.
  • Polypeptides identified by the method discussed herein may be used for immunotherapy by, for example, the cross-linking of monomers to form dimers, trimers, pentamers and other multimers. This may result in better affinity for antigen molecules and slower dissociation rates for some antigens. Another possible approach is to link or fuse polypeptides to a variety of molecules with various functions. For example, antibody fragments may be linked to radionuclides, cytotoxic drugs, toxins, peptides, proteins, enzymes, liposomes, lipids, T-cell superantigens or viruses in order to target and destroy or modify specific cells or molecules.
  • Once the VHs or VLs identified by the selection method described herein have been isolated, they can be further manipulated to select for improved biophysical properties such as solubility, stability, monomericity, binding specificity, human origin or high expressability. This can be achieved by in vitro recombination techniques such as DNA shuffling or a staggered extension process. DNA shuffling involves cutting the nucleic acid sequence of first (donor) and second (acceptor) polypeptides, such as antibody fragments, into random fragments, then reassembling the random fragments by a PCR-like reaction. The reassembled fragments are then screened to select for the desired properties.
  • For example, one or more VHs with high stability (donors) can be mixed with one or more VHs lacking sufficient stability (acceptors) and subjected to DNA shuffling. This generates mutants of the acceptor VHs which have incorporated stability residues from the donor VHs. The newly stable mutants can be identified by the methods described herein, or through other evolutionary protein screening systems such as ribosome display, yeast display, bacterial cell display and phage display. Similarly, this technique can be used to transfer desirable traits such as solubility, monomericity, and high expression.
  • This technique may be used where both donor and acceptor VHs have desirable properties, to produce a VH with both properties. For example, an unstable donor VH which binds to an important therapeutic or diagnostic ligand can be shuffled with a stable acceptor VH. In order to ensure that new generated stable VHs also have the ability to bind to the ligand, the screening system may involve a ligand binding step.
  • DNA shuffling may also be useful for humanizing non-human VHs such as camelid heavy chain antibody variable domains and nurse shark and wobbegong shark variable domains, or non-human VLs which bind to therapeutic targets. Human VHs and VLs with desirable properties such as solubility, stability, monomericity and high expressability may be used as donors. For example, one or more human VHs with good stability (donors) can be mixed with one or more non-human therapeutic VHs (acceptors) and subjected to DNA shuffling. This generates mutants of the acceptor VHs which are both stable and humanized. The newly generated humanized and stable mutants can be identified by the methods described herein, or through other evolutionary protein screening systems such as ribosome display, yeast display, bacterial cell display and phage display. In a further example, the acceptor VH could be a therapeutic VHH (camelid heavy chain antibody variable domain).
  • Further, this technique is also useful for selecting desirable properties of polypeptides other than VHs and VLs. As discussed above, the donor polypeptide and the acceptor polypeptide may be both human, or the donor may be human and the acceptor non-human.
  • A possible approach for imparting solubility, monomericity, high expressability or stability to VHs and VLs may be through grafting complementarity determining regions (CDRs) onto acceptor VHs and VLs. Since CDRs are known to be involved in the solubility and stability of single-domain antibodies, and accordingly the grafting of these regions, such as the CDRs from VHs and VLs isolated by the methods described herein, may impart solubility and/or stability to acceptor VHs and VLs.
  • Human Monomeric VHs and VLs
  • Several monomeric human VHs with different germline and overall sequences were identified (see Figure 1 and SEQ ID NO.8 through 22) from a naïve human VH phage display library by this selection method based on phage plaque size. The VHs remain functional and monomeric following trypsin treatment at 37°C, weeks of incubations at 37°C or months of storage at 4°C, have high thermal refolding efficiencies, are produced in good yields in E. coli and possess protein A binding activity.
  • In addition, several monomeric human VLs were identified (see Figure 6 and SEQ ID NO. 23 through 54). The VLs are also produced in good yields in E. coli and possess protein L binding activity.
  • Such properties will also be manifested by VHs from synthetic libraries that utilize the above VHs as scaffolds. Thus, such libraries may yield therapeutic or diagnostic VHs which would have good efficacy at physiological temperature, extended shelf life and a cost-effective production. High thermal refolding efficiency characteristic would further extend the biotechnological applications of these libraries to situations where VH binders are required to maintain their activity after exposure to transient high temperatures. The VHs should also be very suitable for intrabody applications because of their desirable biophysical properties. The protein A binding property will simplify VH purification and detection in diagnostic tests, immunoblotting and immunocytochemistry and can be exploited to enhance library performance by removing nonfunctional VHs from the libraries. Similarly, libraries that utilize VLs as scaffolds will yield therapeutic or diagnostic VLS which have similarly desirable properties. Since VLs bind with protein L, VL purification and detection is simplified by taking advantage of this protein L binding property.
  • Display libraries built on the present VHs and VLs may also be a useful source of diagnostics and detection agents.
  • Previously reported fully human VHs with favorable biophysical properties were based on a single V germline sequence: DP-47 ((Jespers, L. et al., 2004b;Jespers, L. et al., 2004a). The observation that the monomeric human VHs in this study stem from six different germline sequences including DP-47, demonstrates that stable VHs are not restricted in terms of germline gene usage. In fact, it is very likely that we would have isolated monomeric VHs of family and germline origins different from the ones we describe here had we not restricted our selection to a subset of V H3 family VHs with protein A binding activity. It is not possible to pinpoint amino acid mutations (Table 1) responsible for the observed biophysical behavior of the present VHs due to the occurrence of multiple mutations in VHs and the fact that CDR3 is also known to be involved in shaping the biophysical profiles of sdAbs. It may be, however, that mutations at positions known to be important for sdAbs stability and solubility, eg., V37F in HVHP423 and HVHP44B, or mutations occurring multiple times at the same position, e.g., L5V/Q and V5Q in nine VHs, have a role in determining VHs biophysical properties. In terms of library construction, it would be desirable that the monomericity of the present VHs not be dependent on CDRs, in particular CDR3, so that CDR randomization be performed without the worry of jeopardizing library stability. In this regard, the VHs with smaller CDR3, e.g, HVHB82, may be preferred scaffolds since there would be less dependence on CDR3 for stability.
  • The diversity of the present VHs and VLs in terms of overall sequence and CDR3 length should allow the construction of better-performing libraries. Synthetic VH libraries have been constructed on single scaffolds. Such an approach to repertoire generation is in sharp contrast to the natural, in vivo "approach" which utilizes a multiplicity of scaffolds. Based on the sequences reported here one can take advantage of the availability of the diverse set of VHs and VLs and create libraries which are based on multiple VH and VL scaffolds. Such libraries would be a better emulation of in vivo repertoires and therefore, would have a more optimal complexity. Of the three CDRs in sdAbs, CDR3 generally contribute most significantly to repertoire diversity and for this reason CDR3 randomization on VH and VL scaffolds are typically accompanied by concomitant varying of CDR3 length. While this significantly improves library complexity, it may also compromise library stability by disrupting the length of the parental scaffold CDR3. The heterogeneity of the VHs and VLs disclosed herein in terms of CDR3 length permit the creation of libraries with both good complexity, good stability and good biophysical characteristics. Such libraries would preferably consist of sub-libraries, where each sub-library is created by CDR3 randomization (and CDR1 and/or CDR2 randomization, if desired) on a single VH or VL scaffold without disrupting the parental CDR3 length.
  • The versatility of the present VHs and VLs is also beneficial in terms of choosing an optimal VH or VL framework for humanizing VHHs, VHs and VLs which are specific to therapeutic targets. High affinity camelid VHHs against therapeutic targets can be obtained from immune, non-immunized or synthetic VHH libraries with relative ease and be subsequently subjected to humanization (CDR grafting, resurfacing, deimmunization) to remove possible VHH immunogenicity, hence providing an alternative to human VH library approach for production of therapeutic VHs. Generating high affinity therapeutic VHs by the latter approach may often require additional tedious and time consuming in vitro affinity maturation of the lead binder(s) selected from the primary synthetic human VH libraries.
  • Nonhuman VHs against therapeutic targets can be obtained from immune, non-immunized or synthetic VH libraries with relative ease and be subsequently subjected to humanization (CDR grafting, resurfacing, deimmunization) to eliminate nonhuman VH immunogenicity, hence providing an alternative to human VH library approach for production of therapeutic VHs.
  • Nonhuman VLs against therapeutic targets can be obtained from immune, non-immunized or synthetic VHH libraries with relative ease and be subsequently subjected to humanization (CDR grafting, resurfacing, deimmunization) to eliminate VHH immunogenicity, hence providing an alternative to human VL library approach for production of therapeutic VLs.
  • A number of evolutionary approaches for selection of proteins with improved biophysical properties have been described (Forrer, P. et al., 1999;Waido, G. S., 2003); (Jespers, L. et al., 2004a;Jung, S. et al., 1999;Matsuura, T. et a/., 2003). Typically, stability pressure is required to ensure preferential selection of stable variants over unstable or less stable ones from a library population. For example, in a related work, heat treatment of VH phage display libraries was required to select aggregation resistant VHs (Jespers, L. et al., 2004a). Examples of evolutionary selection approaches involving phage display include conventional phage display, selectively infective phage and the proteolysis approaches. ln the first two approaches affinity selection is used to select stable species from a library, based on the assumption that stable proteins possess better binding properties for their ligand than the unstable ones. However, even with the additional inclusion of a stability selection step, these approaches may primarily enrich for higher affinity rather than for higher stability (Jung, S. et al., 1999). A binding step requirement also limits the applicability of these approaches to proteins with known ligands. The third, proteolysis approach is based on the fact that stable proteins are generally compact and therefore are resistant to proteases whereas the unstable ones are not. The phage display format is engineered in such a way that the protease stability of the displayed protein translates to phage infectivity. Thus, when a variant phage display library is treated with a protease, only the phages displaying stable proteins retain their infectivity and can subsequently be selected by infecting an E. coli host. Since this approach is independent of ligand binding, it has general utility. However, even stable and well folded proteins have protease sensitive sites, e.g., loops and linkers, and this could sometimes hinder the selection of stable species in a proteolysis approach (Bai, Y. et al., 2004).
  • By contrast, in the present evolutionary approach, proteins with superior biophysical properties are simply identified by the naked eye. The approach does not require ligand binding, proteolysis or destabilization steps, and thus, avoids complications which may be encountered in the reported selection approaches. No requirement for a binding step also means that this approach has general utility. As an option, a binding step may be included to ensure that the selected proteins are functional. However, the dependency of the present approach on plating (for plaque visualization) introduces a possible logistical limitation in terms of the number of plates that can be handled and thus limits its application to smaller libraries. Nonetheless, the utility of the current approach can be extended to large libraries, if the library is first reduced to a manageable size. This can be done, for example, by incorporating into the selection system a step which would remove large populations of unstable species, e.g., library adsorption on a protein A surface, or on a hydrophobic interaction column to remove poorly folded proteins with exposed hydrophobic surfaces (Matsuura, T. et al., 2003). Here, the approach was used to select VHs and VLs of good biophysical properties in a background of very unstable VHs and VLs. However, it may be more difficult to select the "best" species from a mutant library which is populated with proteins with reasonably good stabilities. ln this case, the lead variants may be identified based on the rate of plaque formation by using shorter incubation times, or based on plaque size and frequency criteria.
  • The present selection approach can be extended to identification of stable and well-folded antibody fragments such as scFvs and Fabs with the optional inclusion, in the selection system, of a binding step involving protein L, A or any ligand, as well as stable non-antibody scaffolds and variants thereof. Moreover, the observed correlation between phage plaque size and VH expression yield means that one can utilize the present approach for acquiring high-expressing versions of proteins with otherwise poor or unsatisfactory expression from mutant phage display libraries. This application would be particularly appealing in the case of therapeutic proteins or expensive poor-expressing protein reagents where boosting protein expression would significantly offset protein production cost.
  • Binding analyses of pentamers
  • Both VLs and VHs are amenable to pentamerization and the pentamerization can be used to quickly convert a low affinity VL or VH monomer to a high affinity VL or VH pentamer. Such pentamers are invaluable diagnostics and detection agents. ln such applications, the binding of a VL or VH pentamer to its target can be detected by a reporter molecule such as an enzyme (for example, horse radish peroxidase or alkaline phosphatase), or a fluorescent molecule conjugated to the pentamer. Alternatively, the binding of the pentamer can be detected by a secondary molecule which is conjugated to a reporter molecule. The secondary molecule can be specific to the pentamer itself or to a tag thereof, such as a 6His tag or c-Myc tag. For example, a typical secondary molecule is an immunoglobulin.
  • The interactions between the VHs and protein A and VLs with protein L are fundamentally different from those between VHs and VLs with their target antigens. The antigen binding of a VH or a VL involves three antigen binding loops which form the combining site of an antibody domain. The protein A binding of a VH with protein A binding activity and a VL with protein L binding activity involve binding sites and residues on the antibody domains that are totally distinct from the antibody combining site. Thus, a VH with protein A binding activity can simultaneously bind to protein A and its target antigen and a VL with protein L binding activity can simultaneously bind to protein L and its target antigen. Since the present VHs and VLs have affinity for protein A and L, respectively, protein A and L can be used as the secondary molecule for detection and diagnostic applications mentioned above. The human VH and VL pentamers can also be used for therapy.
  • Pathogen detection by the pentamers
  • The protein A and L binding activity of the VHs and VLs can be used to detect bacteria which have protein A and/or L on their surfaces. Protein A is present on the surface of the pathogenic bacteria, Staphylococcus aureus. Thus, the VHs with protein A binding activity such as the ones described here can be used to detect S. aureus. Similarly, the VL monomers and VL Pentamers with protein L binding activity can be used for the detection of bacteria, in particular pathogenic bacteria such as Peptostreptococcus magnus, which have protein L on their cell surface.
  • Protein L is implicated as a virulent factor in the pathogenesis of P. magnus (Ricci, S. et al., 2001) in humans. In vaginosis, protein L is thought to exert its effect by cross-linking surface associated IgE. VL monomers and/or pentamers with protein L binding activity have potential as therapeutics since they could interfere with the IgE cross-linking action of protein L.
  • Protein A is implicated as a virulent factor in the pathogenesis of S. aureus in humans (Fournier, B. et al., 2004). Its virulence has been attributed to its ability to interact with host components including binding to antibodies. VH monomers and/or pentamers with protein A binding activity have potential as therapeutics since they could interfere with the interaction of of protein A with host components.
  • EXAMPLES Identification and sequence analysis of monomeric human VHs
  • During the course of the construction of fully human and Ilaminated human VH libraries, it was learned that the phages displaying monomeric Ilaminated VHs formed larger plaques on bacterial lawns than phages displaying fully human VHs with aggregation tendencies. Thus, plaque size was used as a means of identifying rare, naturally-occurring monomer VHs from the human VH repertoire ( Figure 1 ). To this end, a phage library displaying human VHs with a size of 6 x 108 was constructed and propagated as plaques on agar plates. On the titer plates, the library consisted essentially of small plaques interspersed with some large ones. PCR on twenty clones revealed that the small plaques corresponded to the VH-displaying phages while the large ones represented the wild type phages, i.e., phages lacking VH sequence inserts. None of the VH-displaying phages were found with large plaque morphology. This was not unexpected due to the paucity of the monomeric VHs in the human repertoire and the large size of the library. To facilitate the identification of monomeric VHs, it was decided to reduce the library to a manageable size and remove the interfering wild type phages with large-plaque-size morphology by panning the library against protein A which binds to a subset of human VHs from V H3 family.
  • Following a few rounds of panning, the library became enriched for phage producing large plaques, and PCR and sequencing of more than 110 such plaques showed that all had complete VH open reading frames. The size of the large plaques which were picked for analysis is represented in Figure 1. Sequencing revealed fifteen different VHs which belonged to the V H3 family and utilized DP-38; DP-47, V3-49, V3-53, YAC-5 or 8-1 B germline V segments (Table 1; Figure 2 ). DP-38 and DP-47 germline sequences have been previously implicated in protein A binding. In addition, all VHs had a Thr residue at position 57 ( Figure 2 ), consistent with their protein A binding activity. The most frequently-utilized germline V segment was DP-47 which occurred in over 50% of the VHs, but the most frequent clone (i.e., HVHP428; relative frequency 46%) utilized the V3-49 germline V segment. HVHP429 with a DP-47 germline sequence was the second most abundant VH with a relative frequency of 21% ( Figure 2 ). The VH CDR3 lengths ranged from 4 amino acids for HVHB82 to 16 amino acids for HVHP430 amino acids, with HVHP430 having a pair of Cys residues in CDR3. Amino acid mutations with respect to the parental germline V segment (residues 1-94) and FR4 (residues 103-113) sequences, were observed in all VHs and ranged from two mutations for HVHP44 (L5V and Q105R) and HVHB82 (E1Q and L5Q) to sixteen mutations for HVHP426 (Table 1). Mutations were concentrated in the V segments; only two mutations were detected in all the fifteen FR4s, at positions 105 and 108. HVHP44 and HVHB82 differed from other VHs in that they both had a positively-charged amino acid at position 105 instead of a Gln (Table 1; Figure 2 ). However, while the positively-charged amino acid in HVHP44 was acquired by mutation, the one in HVHB82 was germline-encoded. Except for HVHP423 and HVHP44B, the remaining VHs had the germline residues at the key solubility positions: 37V/44G/45L/47W or 37F/44G/45L/47W (HVHP428); HVHP423 and HVHP44B had a V37F mutation. Mutations at other positions which are shown or hypothesized to be important in VH solubility included seven E6Q, three S35T/H, one R83G and one K83R, one A84P and one T84A and one M108L. Frequent mutations were also observed at positions 1 and 5 which included eleven E1Q, eight L5V/Q and one V5Q mutations.
  • Biophysical characterization of the human VHs
  • All VHs except HVHP44B, which was essentially the same as HVHP423, were expressed in one-litre-culture volumes in E. coli strain TG1 in fusion with c-Myc-His5 tag and purified to homogeneity from periplasmic extracts by immobilized metal affinity chromatography (IMAC). The expression yields ranged from 1.8 to 62.1 mg of purified protein per liter of bacterial culture in shaker flasks with majority of VHs having yields in several milligrams (Table 2). In the instance of HVHP423 and HVHP430, another trial under "apparently" the same expression conditions gave yields of 2.4 and 6.4 mg as opposed to 62.1 and 23.7 mg, respectively. This implies that for many of the VHs described here optimal expression conditions should be achieved, without much effort, resulting in expression yields significantly higher than the reported values in Table 2. As expected, all the VHs bound to protein A in surface plasmon resonance (SPR) analyses, with K D s of 0.2-3 µM, a range and magnitude comparable to the ones reported previously for llama VHH variants with protein A binding activity. None of the VHs bound to the Fab reference surface.
  • The aggregation tendency of the human VHs was assessed in terms of their oligomerization states by gel filtration chromatography and NMR (Table 2). All VHs were subjected to Superdex 75 gel filtration chromatography. Similar to a llama VHH, i.e., H11C7, all VHs gave a symmetric single peak at the elution volume expected for a monomer, and were substantially free of any aggregates (see the example for HVHP428 in Figure 3A . ln contrast, a typical human VH (i.e., BT32/A6) formed considerable amount of aggregates. For three of the VHs, a minor peak with a mobility expected for a VH dimer was also observed. SPR analyses of the minor peaks gave off-rate values which were significantly slower than those for the monomer VHs, consistent with them being dimers. The dimer peak was also observed in the case of the llama VHH, H11C7. The folding and oligomerization states of the VHs at high concentrations were further studied by NMR spectroscopy. As shown in Table II, all the VH proteins studied appeared to be relatively soluble and assumed a well-folded three-dimensional structure. One-dimensonal NMR spectra of the VH fragments (Fig. 3 B) showed structure folds characteristic of VH domains. The state of protein aggregation was also assessed by use of an PFG-NMR diffusion experiment for the HVHP414 fragment and two isoforms, VH14 and VH14-cMyc- with and without the c-Myc sequence, of the HVHP414. VH14 is a modified version of HVHP414 with a c-Myc N132E mutation and with an additional methionine residue at the N-terminus. In brief, the PFG-NMR data (not shown) indicated that all the protein samples had expected monomeric molecular weights even at the relatively high protein concentrations used for NMR experiments.
  • The stability of the VHs was further investigated in terms of their resistance to trypsin at 37°C integrity following long incubations at 37°C. Trypsin cleaves polypeptide amide backbones at the C-terminus of an Arg or a Lys residue. There are 9-13 Arg and Lys residues in the human VHs ( Figure 2 ). There is also an additional Lys residue in the C-terminal c-Myc tag which is susceptible to digestion by trypsin. Figure 4a is an SDS-PAGE analysis of HVHP414 during trypsin digestion. Within 1 h the original band was completely converted to a single product which had a mobility expected for the VH with no c-Myc-HiS5 tag. The same result was obtained for 12 other VHs following a one-hour incubation with trypsin. Mass spectrometry on a randomly selected sample of the trypsin-treated VHs (i.e., HVHP414, HVHP419, HVHP420, HVHP423, HVHP429, HVHP430 and HVHM81) confirmed that in every case the molecular mass of the digested product corresponded to a VH with the c-Myc Lys as the C-terminal residue. HVHM41 gave a significantly shorter fragment than the rest upon digestion, and in this case mass spectrometry experiments mapped the cleavage site to the Arg99 in CDR3 (data not shown).
  • Eleven VHs ranging in concentration from 0.32 mg/ml (HVHP428) to 3.2 mg/ml (HVHP420) were incubated at 37°C for 17 days. Their stability was subsequently determined in terms of oligomerization state and protein A binding. As shown by gel filtration chromatography, treatment of VHs at 37°C did not induce any aggregate formation: all VHs gave chromatogram profiles which were virtually identical to those of untreated VHs and stayed essentially as monomers (see the example for HVHP420; Figure 4c ). To ensure that the VHs maintained their native fold following 37°C treatment, two VHs, namely, HVHP414 (1.2 mg/ml) and HVHP420 (3.2 mg/ml), were selected at random and their KDs of binding to protein A were determined by SPR (Data shown for HVHP420; Figure 4c inset) and compared to the KDs obtained for untreated VHs (Table 2). The calculated KDs for the heat-treated VHs were 1.4 µM and 1.0 µM for HVHP414 and HVHP420, respectively. These values are essentially identical to the corresponding values for the untreated VHs (Table 2), demonstrating that 37°C treatment of VHs did not affect their native fold. The possibility that VHs may have been in a less compact, non-native fold during the 37°C-incubation periods and resumed their native fold upon returning to room temperature during gel filtration and SPR experiments is unlikely in light of the fact that the VHs were resistant to trypsin at 37°C (see above), a property typically associated for well folded native proteins.
  • The refolding efficiency (RE) of the human VHs was investigated by comparing the KD s of the binding of the native (KDn) and heat-treated, refolded (KDref ) VHs to protein A (Tanha, J. et al., 2002). When a fraction of the VH is inactivated by heat treatment the measured KD would be higher, since this parameter is based on the concentration of folded, i.e., active, antibody fragment. Thus, the ratio of KDn to KDref gives a measure of VH RE. Figure 5 compares sensorgrams for HVHP423 binding to immobilized protein A in native (thick lines) and refolded (thin lines) states at several selected VH concentrations. As can be seen, binding of the refolded VH to protein A is less in all instances, indicating that the unfolding is not fully reversible. For each of the 14 VHs, protein A binding in both native and refolded states was measured at several concentrations, and the KD s and subsequently REs were determined (Table 2; KDref values are not shown). The K Ds and REs of two anti-idiotypic llama VHHs, H11 F9 and H11 E32, which were used as references, were also determined. Four VHs had REs in the range of 92%-95%, similar to the REs for H11F9 and H11B2, 95% and 100%, respectively. Another five had REs in the range of 84%-88% and three over 70%. Only two had significantly lower RE: HVHP413 (52%) and HVHP421 (14%). Several published VHHs examined previously had RE around 50% (van der Linden, R. H. et al., 1999).
  • Human VH phage display library construction and panning. cDNA was synthesized from human spleen mRNA (Ambion Inc., Austin, TX) using random hexanucletide primers and First Strand cDNA™ kit (GE Healthcare, Baie d'Urfé, QC, Canada). Using the cDNAs as template, VH genes with flanking CH sequences were amplified by polymerase chain reaction (PCR) in nine separate reactions using VH framework region 1 (FR1 )-specific primers and an immunoglobin M-specific primer (de Haard, H. J. et al., 1999). The products were get-purified and used as the template in the second round of PCR to construct VH genes using the FR1- and FR4-specific primers (de Haard, H. J. et al., 1999) that also introduced flanking Apal I and Not I restriction sites for cloning purposes. The resultant VH repertoire DNAs were cloned into fd-tetGlllD phage vector and a VH phage display library was constructed (Tanha, J. et al., 2001). Panning against protein A (Amersham Biosciences Inc.) was performed as described (Tanha, J. et al ., 2001). Germline sequence assignment of the selected VHs was performed using DNAPLOT software Version 2.0.1 and V BASE version 1.0 (http://vbase.dnaplot.de/cgi-bin/vbase/vsearch.pl).Llama VHHs H11C7, H11 F9 and H11 B2 were isolated from a llama VHH phage display library by panning against H11 scFv as described (Tanha, J. et al., 2002).
  • VH expression and purification. VHs were cloned into pSJF2 expression vectors by standard cloning techniques (Sambrook, J. Fritsch E. F. and Maniatis T, 1989). Periplasmic expression of sdAbs and subsequent purification by immobilized metal affinity chromatography (IMAC) were performed as described (Muruganandam, A. et al., 2002). Protein concentrations were determined by A280 measurements using molar absorption coefficients calculated for each protein (Pace, C. N. et al., 1995). Gel filtration chromatography of the purified VHs was performed on a Superdex 75 column (GE Healthcare) as described (Deng, S. J. et al., 1995).
  • Binding and refolding efficiency experiments. Equilibrium dissociation constants (KDs) and refolding efficiencies (REs) of VHsN HHs were derived from surface plasmon resonance (SPR) data collected with BIACORE 3000 biosensor system (Biacore Inc., Piscataway, NJ). To measure the binding of VHs to protein A, 2000 resonance units (RUs) of protein A or a reference antigen-binding fragment (Fab) were immobilized on research grade CM5 sensor chips (Biacore Inc.). Immobilizations were carried out at concentrations of 25 µg/ml (protein A) or 50 µg/ml (Fab) in 10 mM sodium acetate buffer pH 4.5, using the amine coupling kit provided by the manufacturer. To measure the binding of the anti-idiotypic llama VHHs to H11 scFv, 4100 RUs of 50 µg/ml H11 scFv or 3000 RUs of 10 µg/ml Se155-4 IgG reference were immobilized as described above. In all instances, analyses were carried out at 25°C in 10 mM HEPES, pH 7.4, containing 150 mM NaCl, 3 mM EDTA and 0.005% P20 at a flow rate of 40 µl/min, and surfaces were regenerated by washing with the running buffer. To determine the binding activities of the refolded proteins, VHs or VHHs were denatured by incubation at 85°C for 20 min at 10 µg/ml concentrations. The protein samples were then cooled down to room temperature for 30 min to refold and were subsequently centrifuged in a microfuge at 14,000 rpm for 5 min at room temperature to remove any protein precipitates. The supernatants were recovered and analyzed for binding activity by SPR as described above. For both folded and refolded proteins data were fit to a 1:1 interaction model simultaneously using BlAevaluation 4.1 software (Biacore Inc.) and KD s were subsequently determined. REs were determined from RE = K D n K D ref × 100
    Figure imgb0001
  • Where KDn is the KD of the native protein and KDref is the KD of the refolded protein.
  • Tryptic digest experiments. 3 µl of a freshly prepared 0.1 µg/µl sequencing grade trypsin (Hoffmann-La Roche Ltd., Mississauga, ON, Canada) in 1 mM HCI was added to 60 µg VH in 100 mM Tris-HCI buffer pH 7.8. Digestion reactions were carried out in a total volume of 60 µl for 1 h at 37°C and stopped by adding 5 µl of 0.1 µg/µl trypsin inhibitor (Sigma, Oakville, ON, Canada). Following completion of digestion, 5 µl was removed and analyzed by SDS-PAGE; the remaining was desalted using ZipTipc4 (Millipore, Nepean, ON, Canada), eluted with 1% acetic acid in 50:50 methanol:water and subjected to VH mass determination by MALDI mass spectrometry.
  • Protein stability studies at 37°C. Single-domain antibodies (sdAbs) at 0.32-3.2 mg/ml concentrations were incubated at 37°C in PBS buffer for 17 days. Following incubation, the protein samples were spun down in a microfuge at maximum speed for 5 min even in the absence of any visible aggregate formation. The samples were then applied onto a Superdex 75 size exclusion column (GE Healthcare) and the monomeric peaks were collected for SPR analysis against protein A. SPR analyses were performed as described above except that 500 RUs of protein A or reference Fab was immobilized and that immobilizations were carried out at concentration of 50 µg/ml.
  • NMR experiments - VH samples for NMR analysis were dissolved in 10 mM sodium phosphate, 150 mM NaCI, 0.5 mM EDTA, and 0.02% NaN3 at pH 7.0. The protein concentrations were 40 µM - 1.0 mM. All NMR experiments were carried out at 298 K on a Bruker Avance-800 or a Bruker Avance-500 NMR spectrometer. One-dimensional (1D) 1H NMR spectra were recorded with 16,384 data points and the spectral widths were 8,992.81 Hz at 500 MHz and 17,605.63 Hz at 800 MHz, respectively. Two-dimensional 1H-1H NOESY spectra of 2,048 x 400 data points were acquired on a Bruker Avance-800 NMR spectrometer with a spectral width of 11,990.04 Hz and a mixing time of 120 ms. In all NMR experiments, water suppression was achieved using the WATERGATE method implemented through the 3-9-19 pulse train (Piotto, M. et al. , 1992;Sklenar, V. et al., 1993). NMR data were processed and analyzed using the Bruker XWINNMR software package. All PFG-NMR diffusion measurements were carried out with the water-suppressed LED sequence (Altieri, A. S. et al., 1995), on a Bruker Avance-500 NMR spectrometer equipped with a triple-resonance probe with three-axis gradients. One-dimensional proton spectra were processed and analyzed using Bruker Xwinnmr software package. NMR signal intensities were obtained by integrating NMR spectra in the methyl and methylene proton region (2.3ppm to -0.3ppm) where all NMR signals were attenuated uniformly at all given PFG strengths.
  • Human VL phage display library construction and panning. cDNAs were synthesized from human spleen mRNA as described above for the human VHs. The cDNA was used as template in PCR to amplify VL genes in 50 µl reaction volumes using six Vκ back primers, 11 Vλ back primers (de Haard, H. J. et al., 1999). four Vκ For primers and two Vλ For primers (Sblattero, D. et al., 1998). The back and forward primers were modified to have flanking Apa Ll and Not l restriction sites, respectively, for subsequent cloning purposes. Forward primers were pooled together in ratios which reflected their degree of degeneracy. Vλ genes were PCRed in 11 separate reactions using the pooled Vλ For primers and 11 individual Vλ back primers. Similarly, Vλ genes were amplified in 6 separate reactions using the pooled Vκ For primers and 6 individual Vλ back primers. The PCR products were pooled, gel purified and digested with Apa LI and Not I restriction endonucleases. The library was constructed as described for human VHs. Plaque PCR was performed on individual library colonies and the amplified VL genes were sequenced as described (Tanha, J. et al., 2003). Panning against protein L (Biolynx Inc., Brockville, ON, Canada) and germline sequence assignment of the selected VLs were performed as described above for human VH library.
  • VL expression and purification. VL expression, purification, concentration determination and gel filtration chromatography were carried out as described for VHs in "VH expression and purification,".
  • Expression and purification of VL and VH pentamers. Specific primers were used in a standard PCR to amplify HVHP328 VH and HVLP335 VL genes. Standard cloning techniques were used to clone the HVHP328 and HVLP335 genes in fusion with VT1B pentamerization domain gene in an expression vector to yield HVHP328PVT2 and HVLP335PTV2 pentamers, (Zhang, J. et al., 2004). Pentamers were expressed and purified as described (Zhang, J. et al., 2004). Protein concentrations were determined as above.
  • Surface plasmon resonance of VLs. The binding kinetics for the interaction of the VLs to protein L were determined by SPR using BIACORE 3000 biosensor system (Biacore, inc., Piscataway, NJ). 680 RUs of protein L or 870 RUs of a Fab reference were immobilized on research grade CM5 sensor chips (Biacore). Immobilizations were carried out at a protein concentration of 50 µg/ml in 10 mM acetate buffer pH 4.5 using the amine coupling kit supplied by the manufacturer. All measurements were carried out at 25°C in 10 mM HEPES buffer pH 7.4, containing 150 mM NaCl, 3 mM EDTA and 0.005% P20 at a flow rate of 50 µl/min or 100 µl/min. Surfaces were regenerated by washing with the running buffer. Data were evaluated using the BlAevaluation 4.1 software (Biacore, Inc.).
  • Surface plasmon resonance of the pentameric VL and VH. The binding kinetics for the interaction of HVHP328PVT2 with protein A and HVLP335PTV2 with protein L were also determined by SPR. 520 RUs of protein A or a Fab reference were immobilized as above. For the VL pentamer, the same surfaces prepared above were used. Measurements were carried out as above but at a flow rate of 20 µl/min. Surfaces were regenerated by washing with 50 mM HCl for 3 s. Data were evaluated as described for the monomers.
  • Cell microagglutination
  • A single S. aureus colony from a BHI plate was used to inoculate 15 mL of BHI media. The bacteria were grown overnight at 37°C at 200 rpm. In the morning, the culture was spun down in a swinging bucket, Sorvall RT6000B refrigerated centrifuge at 4000 rpm for 10 min, the supernatant was removed and the cell pellet was re-suspended in PBS buffer. The cells were re-spun, the supernatant was removed and the cell pellet was re-suspended again in PBS buffer. The cells were diluted to an A600 of 1.0, and serial dilutions of the cells were spread on BHl plates at 37°C for overnight growth. The cell titer was determined in the morning. An A600 of 1.0 corresponded to 1.5x 109 cells ml1. Identical steps were taken to prepare E. coli starin TG1 cells for subsequent microagglutination assays, except that the growth media was 2xYT. The viable counts were similar, A6001.0 = 2.1x109 cells ml-1.
  • To perform microagglutination assays, two fold dilutions of HVHP328PVT2 in PBS were performed from wells 1 to 11 in a microtiter plate. Well 12 (blank) had only PBS. The total volume in each well was 50 µl. Subsequently, 1 x 108 S. aureus cells in 50 µl PBS was added to all wells and the plate was incubated overnight at 4°C. To have a permanent record of the results, a picture was taken from the plate in the morning. For the pentamer control experiment, HVHP328PVT2 was replaced with the VL pentamer, HVLP335PTV2. In the cell control experiments, the same two sets of experiments were repeated with E. coli TG1 cells.
  • Identification and sequence analysis of monomeric human VLs
  • Essentially the same selection method which was employed to isolate soluble VHs from a human VH phage display library was applied to a human VL library for isolating soluble, monomeric VLs. A human VL library with a size of 3 x 106 was constructed. Twenty four plaques from the library titer plates were picked and their VL genes were PCRed and sequenced. The sequences were diverse in terms of germ-line origin although 75% of the VLs were of Vλ origin (data not shown). Three rounds of panning against protein L resulted in To perform microagglutination assays, two fold dilutions of HVHP328PVT2 in PBS were performed from wells 1 to 11 in a microtiter plate. Well 12 (blank) had only PBS. The total volume in each well was 50 µl. Subsequently, 1 x 108 S. aureus cells in 50 µlPBS was added to all wells and the plate was incubated overnight at 4°C. To have a permanent record of the results, a picture was taken from the plate in the morning. For the pentamer control experiment, HVHP328PVT2 was replaced with the VL pentamer, HVLP335PTV2. In the cell control experiments, the same two sets of experiments were repeated with E. coli TG1 cells.
  • Identification and sequence analysis of monomeric human VLs
  • Essentially the same selection method which was employed to isolate soluble VHs from a human VH phage display library was applied to a human VL library for isolating soluble, monomeric VLs. A human VL library with a size of 3 x 106 was constructed. Twenty four plaques from the library titer plates were picked and their VL genes were PCRed and sequenced. The sequences were diverse in terms of germ-line origin although 75% of the VLs were of Vλ origin (data not shown). Three rounds of panning against protein L resulted in enrichment for large plaques. Thirty-nine of large plaques were sequenced and 32 unique sequences were identified (Figure 6). HVLP325, HVLP335 and HVLP351 occurred at frequency of 3, 4 and 2, respectively. Except for HVLP389 which is of lambda class (subgroup Vλ1, germline 1b), the remaining 31 VLs belonged to the Vκ class. Of the 31 kappa VLs, 24 fall within the Vκlll subgroup and 7 within the Vκ1 subgroup. Sixteen of the 24 Vκlll sequences utilize L6 germline sequence with the remaining utilizing A27, L2 and L6 germline sequences. The Vκ1 subgroup VLs are originated from 02/012 or A30 germline sequence. Noticeable mutations occurred at position 96. The germline amino acids at this position are aromatic and hydrophobic amino acids Trp, Phe, Tyr, Leu or Ile for kappa VLs and Tyr, Val or Ala for lambda VLs. But in the selected pool of kappa VLs only 5 out of 31 have their germline amino acids at position 96: HVLP325, HVLP349, HVLP388, HVLP3109 and HVLP393. 21 amino acids at position 96 are charged of which 20 are positively-charged: Arg, Lys or His. Two amino acids are Pro, one Gin, one Ser and one Thr. Of seven kappa VLs analyzed by gel filtration chromatography for monomericity, six which had Arg or Lys at position 96 were also monomers, whereas HVLP325 with the germline amino acid Leu at position 96 formed aggregates (see below). Similarly, HVLP389 which was of the lambda class and had a germline mutation to Ser was also monomeric (see below). These data correlates the deviation from the germline amino acids at position 96 (27 out of 32) with improved biophysical properties of VLs such as monomericity.
  • Eighteen VLs of the kappa class had their last three residues (105-107) replaced with amino acids Thr, Val and Leu which are only found in lambda VLs. These substitutions may have had a role in improving the biophysical properties of the kappa VLs, resulting in the selection of the aforementioned VLs over the parental clones with the original kappa residues at position 105-107.
  • Characterization of the human VLs
  • Eight of the selected VLs with different V germline origins were expressed in E. coli in one-liter cultures and purified: HVLP324, HVLP325, HVLP335, HVLP342, HVLP351, HVLP364, HVLP389 and HVLP3103 (Table 6). All were expressed in good yields ranging from 6.2 mg for HVLP324 to around 75 mg for HVLP335 and HVLP364.
  • The aggregation tendency of the human VLs was assessed in terms of their oligomerization state by gel filtration chromatography. VLs were subjected to Superdex 75 gel filtration chromatography at a concentration of 0.6 mg/ml. All except HVLP325 were essentially free of aggregates and gave symmetric single peaks with the mean apparent molecular mass of 12.7 kDa (range, 6.2-19.2 kDa) ( Figure 7A and Table 3). This is in agreement with the expected molecular mass for monomeric VLs, 13.4-13.8 kDa. Variation in apparent molecular mass for single-domain antibodies has been reported previously (Jespers, L. et al., 2004a; (Stevens, F. J. et al., 1980). For HVLP325, the aggregates formed 11% of the total protein (aggregate plus monomer). HVLP351, HVLP342, HVLP335 and HVLP3103, were still monomer when tested at their highest concentration available, i.e., 0.89 mg/ml 1.0 mg/ml, 4.9 mg/ml and 5.9 mg/ml, respectively ( Figure 7B )
  • VLs were subjected to Superdex-75 chromatography prior to BIACORE analysis and purified monomer peaks collected even in the absence of any evidence of aggregated material. In SPR analysis, all selected VLs bound to protein L (Figure 8). This was not unexpected since the VLs were isolated by panning against protein L. For all, the KDs of binding to protein L were in 0.6-3 µM (Table 3). HVLP324 and HVLP342 had additional smaller KDs, 10 nM and 40 nM, respectively. Low affinity and high affinity bindings of VLs of Vκl subgroup to protein L have been reported previously (Reference). Both, HVLP324 and HVLP342, belong to Vκl subgroup (Table 3). As expected, the kinetic and equilibrium data were consistent with the monomeric peak being indeed monomeric.
  • Binding analyses of pentamers
  • Bindings of HVHP328PVT2 pentamer to protein A and HVLP335PTV2 pentamer to protein L were determined by surface plasmon resonance ( Figure 9 ).The association rates were independently calculated from plots of kobs versus concentration. More than one dissociation rate (k d ) could be calculated due to the heterogeneity in multivalent binding amongst the pentamer population. Therefore, more than one equilibrium dissociation constant, KD, could be obtained. HVHP328PTV2 and HVLP335PTV2 had minimum KDs of 2 nM and 200 pM, respectively (Table 4). With slower k d s, HVHP328PTV2 and HVLP335PTV2 had KDs as low as 900 and 90 pM, respectively.
  • Pathogen detection by VLs and VHs
  • The protein A and L binding activity of the VHs and VLs can be used to detect bacteria which have protein A and/or L on their surfaces. This is possible if the VHs and VLs are soluble and monomeric (lack of tendency to aggregate) such as the VHS and VLs here. Variable domains derived from antibodies which lack light chains such as camelid heavy chain antibodies or nurse shark and wobbegong shark lgNARs are naturally soluble and monomeric. From these, those with protein A and L binding activity.can also be used to detect bacteria which have protein A and/or L on their surfaces. Protein A is present on the surface of the pathogenic bacteria, Staphylococcus aureus. Thus, the VHS with protein A binding activity such as the ones described here can be used to detect S. aureus. We performed a microagglutination assay to detect the ability of HVHP328PVT2 VH pentamer to bind to S. aureus. A constant number of bacterial cells were incubated with two-fold dilutions of HVHP328PVT2 in microtiter wells (wells 1-11) ( Figure 10 ). Well 12 had buffer instead of the pentamer. lf the VHS bind to the bacterial cells, then the pentamer because of its multimeric nature should be able to cross-link the cells and results in cell agglutination. The agglutinated cells will appear as diffused cells in a microtiter well ( Figure 10 ). ln the absence of any binding, no agglutination should occur, hence no agglutination, and the cells will appear as a dot at the bottom of the well. As shown in Figure 10, the pentamer binds to the S. aureus, since there is agglutination of cells. The agglutination is observed up to well 7. Beyond well 7 the concentration of the pentamer is too low for binding, hence no agglutination. The control VL pentamer does not show any agglutination, demonstrating the specificity of the VH pentamer to S. aureus ( Figure 10 ). The binding is also cell-specific since the VH pentamer as expected does not agglutinate E. coli (TG1 strain) or Salmonella cells (data not shown). Similarly, the VL monomers and VL pentamers with protein L binding activity can be used for the detection of bacteria, in particular pathogenic bacteria such as Peptostreptococcus magnus, which have protein L on their cells surface.
  • It is understood that the examples described above in no way serve to limit the true scope of this invention, but rather are presented for illustrative purposes.
  • Reference List
    • Bai, Y. and Feng, H. (2004). Selection of stably folded proteins by phage-display with proteolysis. Eur.J.Biochem. 271: 1609-1614.
    • Davies, J. and Riechmann, L. (2-21-1994). 'Camelising' human antibody fragments: NMR studies on VH domains. FEBS Lett 339: 285-290.
    • Davies, J. and Riechmann, L. (1995). Antibody VH domains as small recognition units. Biotechnology N.Y. 13: 475-479.
    • de Haard, H. J., van Neer, N., Reurs, A., Hufton, S. E., Roovers, R. C., Henderikx, P., de Bruine, A. P., Arends, J. W., and Hoogenboom, H. R. (6-25-1999). A large non-immunized human Fab fragment phage library that permits rapid isolation and kinetic analysis of high affinity antibodies. J.Biol.Chem. 274: 18218-18230.
    • Deng, S. J., MacKenzie, C. R., Hirama, T., Brousseau, R., Lowary, T. L., Young, N. M., Bundle, D. R., and Narang, S. A. (5-23-1995). Basis for selection of improved carbohydrate-binding single-chain antibodies from synthetic gene libraries. Proc.Natl.Acad.Sci U.S.A 92: 4992-4996.
    • Forrer, P., Jung, S., and Pluckthun, A. (1999). Beyond binding: using phage display to select for structure, folding and enzymatic activity in proteins. Curr.Opin.Struct.Biol. 9: 514-520.
    • Foumier, B. and Klier, A. (2004). Protein A gene expression is regulated by DNA supercoiling which is modified by the ArlS-ArlR two-component system of Staphylococcus aureus. Microbiology 150: 3807-3819.
    • Hamers, C. C., Atarhouch, T., Muyldermans, S., Robinson, G., Hamers, C., Songa, E. B., Bendahman, N., and Hamers, R. (6-3-1993). Naturally occurring antibodies devoid of light chains. Nature 363: 446-448.
    • Jespers, L., Schon, O., Famm, K., and Winter, G. (2004a). Aggregation-resistant domain antibodies selected on phage by heat denaturation. Nat.Biotechnol. 22: 1161-1165.
    • Jespers, L., Schon, O., James, L. C., Veprintsev, D., and Winter, G. (4-2-2004b). Crystal Structure of HEL4, a Soluble, Refoldable Human V(H) Single Domain with a Germ-line Scaffold. J.Mol.Biol. 337: 893-903.
    • Jung, S., Honegger, A., and Pluckthun, A. (11-19-1999). Selection for improved protein stability by phage display. J.Mol.Biol. 294: 163-180.
    • Matsuura, T. and Pluckthun, A. (3-27-2003). Selection based on the folding properties of proteins with ribosome display. FEBS Lett. 539: 24-28.
    • Muruganandam, A., Tanha, J., Narang, S., and Stanimirovic, D. (2002). Selection of phage-displayed llama single-domain antibodies that transmigrate across human blood-brain barrier endothelium. FASEB J. 16: 240-242.
    • Pace, C. N., Vajdos, F., Fee, L., Grimsley, G., and Gray, T. (1995). How to measure and predict the molar absorption coefficient of a protein. Protein Sci. 4: 2411-2423.
    • Ricci, S., Medaglini, D., Marcotte, H., Olsen, A., Pozzi, G., and Bjorck, L. (2001). Immunoglobulin-binding domains of peptostreptococcal protein L enhance vaginal colonization of mice by Streptococcus gordonii, Microb.Pathog. 30: 229-235.
    • Sambrook, J. F. E. F. a. M. T. (1989). "Molecular Cloning: A laboratory Manual (2nd ed.)", Cold Spring Harbor Laboratory, Cold Spring Harbor, NY..
    • Sblattero, D. and Bradbury, A. (1998). A definitive set of oligonucleotide primers for amplifying human V regions. Immunotechnology. 3: 271-278.
    • Tanha, J., Dubuc, G., Hirama, T., Narang, S. A., and MacKenzie, C. R. (5-1-2002). Selection by phage display of llama conventional V(H) fragments with heavy chain antibody V(H)H properties. J.Immunol.Methods 263: 97-109.
    • Tanha, J., Muruganandam, A., and Stanimirovic, D. (2003). Phage Display Technology for Identifying Specific Antigens on Brain Endothelial Cells. Methods Mol.Med. 89: 435-450.
    • Tanha, J., Xu, P., Chen, Z. G., Ni, F., Kaplan, H., Narang, S. A., and MacKenzie, C. R. (7-6-2001). Optimal design features of camelized human single-domain antibody libraries. J.Biol.Chem 276: 24774-24780.
    • van der Linden, R. H., Frenken, L. G., de Geus, B., Harmsen, M. M., Ruuls, R. C., Stok, W., de Ron, L., Wilson, S., Davis, P., and Verrips, C. T. (4-12-1999). Comparison of physical chemical properties of llama VHH antibody fragments and mouse monoclonal antibodies. Biochim.Biophys.Acta 1431: 37-46.
    • Waldo, G. S. (2003). Genetic screens and directed evolution for protein solubility. Curr.Opin.Chem.Biol. 7: 33-38.
    • Ward, E. S., Gussow, D., Griffiths, A. D., Jones, P. T., and Winter, G. (10-12-1989). Binding activities of a repertoire of single immunoglobulin variable domains secreted from Escherichia coli [see comments]. Nature 341: 544-546.
    • Zhang, J., Li, Q., Nguyen, T. D., Tremblay, T. L., Stone, E., To, R., Kelly, J., and MacKenzie, C. R. (7-30-2004). A pentavalent single-domain antibody approach to tumor antigen discovery and the development of novel proteomics reagents. J.Mol.Biol. 341: 161-169.
    Table 1. VH sequence deviations from parental germline sequences
    VH V/J germlines Amino acid deviation from V and FR4 germline sequences
    HVHP44 DP47/JH4b L5V.Q105R
    HVHBB2 DP47/JH6c E1Q.L5Q
    HVHP421 DP47/JH4b E1Q, V2L. L5Q. L11V. G16R
    HVHP419 DP47/JH4b E1Q, V2L, L5Q, T77S, R83G,K94R
    HVHP430 DP47/JH3b E1Q. L5V, V121,Q13K, S31N, G52AS. L78V, A93V, K94R
    HVHP429 DP47/JH4 L5V, G10T, S301, S31N. G42D, E46D, A50T, G52aN, S53N, S56A K75N, A84P, E85D
    HVHM41 DP47/JH3a E1Q, L5V. E6Q, G16R, T28A, S53G, G55D, S56H, M108L
    HVHM81 DP47JH3a L5V, E6Q.G16R, S30D, S31D, S35H, A50G, G55A, E85G, V89L. K94R
    HVHP428 V3-49/JH4b E1Q. V2L, V5Q. R16G. T23A, G30S, D31S, T60A, G73D, K83R, T84A, V89M, T93A
    HVHP420 DP-38/JH4b E1Q. S35T, S52aT
    HVHP414 DP-38/JH3b E1D. E6Q. A23T, T28P, K52T, A60V
    HVHP423 V3-53/JH1 E1Q, V2M, E6Q, L11V, 112V, N32S, Y33R, V37F, K43M, K64R, T68S, V89L
    HVHP44B V3-53/JH1 E1Q, E6Q,N32S, Y33R, V37F, K43M, Y58S, K64R, T68S. V89L
    HVHP413 YAC-5/JH3b E1Q, E6Q, Q13K, V29F, S31D, N32Y, V50F
    HVHP426 8-1B/JH3b E1Q. E6Q, L11V, G16R, T281, S30D, S31G, N32Y, Y33A, S35H, K43Q. 151T, Y52N, S53N, Y58S, L78V
    Table 2. Biophysical characteristics of the human VHS
    VH/VHH Exp.# (mg) KD (µM) Trypsin resistance RE (%)
    HVHP44 8.2 1.3 93
    HVHB82 5.9 0.2 71
    HVHP421 5.5 1.0 14
    HVHP419 3.4 1.6 84
    HVHP430 6.4,23.7 2.3 88
    HVHP429 3.4 1.3 86
    HVHM41 1.8 0.5 92
    HVHM81 4.3 1.3 87
    HVHP428 3.1 1.8 95
    HVHP420 59.0 1.2 92
    HVHP414 11.8 1.6 73
    HVHP423 2.4,62.1 3.0 86
    HVHP413 5.8 0.3 52
    HVHP426 6.3 0.8 70
    H11F9* ND 3.5 ND 95
    H11B2* ND 2.0 ND 100
    #expression yield per liter of bacterial culture
    * K D s and RE s were determined against H 11 scFv.
    Table 3. Characteristics of the human VLS
    VL Subgroup Expressiona KD Oligomerization stateb
    mg µM
    HVLP324 VKl 6.9 0.2, 0.01c Monomer
    HVLP325 VKlll 6.2 1 Monomer/Aggregate
    HVLP335 Vklll 73.5 2 Monomer
    HVLP342 VKl 7.7 0.6, 0.04c Monomer
    HVLP351 VKlll 8.9 2 Monomer
    HVLP364 Vklll 77.1 1 3 Monomer
    HVLP389 Vλ1 16.7 1 Monomer
    HVLP3103 VKlll 19.0 1 Monomer
    a Expression yield per liter of bacterial culture.
    b Oligomerization state was determined by gel filtration chromatography.
    c The smaller KD values correspond to the binding of the of HVLP324 and HVLP342 to the high affinity sites on protein L.
    Table 4. Kinetic and equilibrium constants for the bindings of HVHP328PTV2 and HVLP335PTV2 to protein A and L, respectively
    Pentabody HVHP328PTV2 HVLP335PTV2
    ka (M-1S-1) 4.3 x 105 1.7 x 106
    kd (S-1) < 1 x 10-3 < 4 x 10-4
    KD (M) < 2 x 10-9 < 2 x10-10
  • The following are embodiments of the invention:
  • Embodiment 1 .
  • A method of identifying target polypeptides, comprising:
    1. (a) obtaining a phage display library capable of expressing a variety of polypeptide sequences;
    2. (b) allowing infection of a bacterial lawn by the library phage; and
    3. (c) identifying phage which form larger than average plaques on the bacterial lawn.
    Embodiment 2.
  • A method as described in embodiment 1, where the target polypeptides are soluble.
  • Embodiment 3.
  • A method as described in embodiment 1, where the target polypeptides are monomeric.
  • Embodiment 4.
  • A method as described in embodiment 1, where the target polypeptides are non-aggregating.
  • Embodiment 5.
  • A method as described in embodiment 1, where the target polypeptides are highly expressed.
  • Embodiment 6.
  • A method as described in embodiment 1, where the target polypeptides are stable.
  • Embodiment 7.
  • A method as described in embodiment 1, where the phage is a filamentous phage.
  • Embodiment 8.
  • A method as described in embodiment 7, where the phage is M13 or fd.
  • Embodiment 9.
  • A method as described in embodiment 6, wherein the stable polypeptides have relatively high thermal refolding efficiency.
  • Embodiment 10.
  • A method as described in embodiment 6, wherein the stable polypeptides have a relatively high melting temperature.
  • Embodiment 11.
  • A method as described in embodiment 6, wherein the stable polypeptides have maintained their functionality following long incubation at 37°C.
  • Embodiment 12.
  • A method as described in embodiment 6, wherein the stable polypeptides are relatively resistant to chemical denaturants.
  • Embodiment 13.
  • A method as described in embodiment 6, wherein the stable polypeptides are relatively resistant to proteases.
  • Embodiment 14.
  • A method as described in embodiment 6, wherein the stable polypeptides have a relatively long shelf life at or above room temperature, at 4°C, or below 0°C.
  • Embodiment 15.
  • A method as described in embodiment 6, wherein the stable polypeptides are functional in intracellular environments.
  • Embodiment 16.
  • A method as described in embodiment 6, wherein the stable polypeptides are functional when administered internally to humans.
  • Embodiment 17.
  • A method as described in embodiment 1, further comprising the steps of:
    • (d) isolating the larger plaque phage in step (c); and
    • (e) determining the sequence or other characteristics of the polypeptides expressed by the larger plaque phage.
    Embodiment 18.
  • A method as described in embodiment 1, wherein the target polypeptides are antibodies or fragments of antibodies.
  • Embodiment 19.
  • A method as described in embodiment 18, wherein the target polypeptides are human VH or VL antibody fragments.
  • Embodiment 20.
  • A method as described in embodiment 1, wherein the target polypeptides are vaccines.
  • Embodiment 21.
  • A method as described in embodiment 1, wherein the target polypeptides are therapeutic proteins.
  • Embodiment 22.
  • A method as described in embodiment 1, wherein the target polypeptides are selected from the group consisting of: single-chain T-cell receptors, T-cell receptor domains, transferin, lipocalins, kunitz domains, ankyrin repeats, and cytotoxic T-lymphocyte-associated antigen.
  • Embodiment 23.
  • A method as described in embodiment 1, wherein the target polypeptides are proteinaceous diagnostic and biochemical reagents.
  • Embodiment 24.
  • A polypeptide having an amino acid sequence selected from the group consisting of: SEQ lD NO:8-54.
  • Embodiment 25.
  • A polypeptide having an amino acid sequence that is substantially identical to the amino acid sequence described in embodiment 24.
  • Embodiment 26.
  • A nucleic acid sequence that encodes a polypeptide as described in embodiment 24 or 25.
  • Embodiment 27.
  • A polypeptide comprising the CDR3 portion of an amino acid sequence as described in embodiment 24.
  • Embodiment 28.
  • A nucleic acid sequence that encodes a polypeptide as described in embodiment 27.
  • Embodiment 29.
  • A polypeptide comprising the FR1 1 portion of an amino acid sequence as described in embodiment 24.
  • Embodiment 30.
  • A nucleic acid sequence that encodes a polypeptide as described in embodiment 29.
  • Embodiment 31.
  • A polypeptide comprising the FR2 portion of an amino acid sequence as described in embodiment 24.
  • Embodiment 32.
  • A nucleic acid sequence that encodes a polypeptide as described in embodiment 31.
  • Embodiment 33.
  • A polypeptide comprising the FR3 portion of an amino acid sequence as described in embodiment 24.
  • Embodiment 34.
  • A nucleic acid sequence that encodes a polypeptide as described in embodiment 33.
  • Embodiment 35.
  • A polypeptide comprising the FR4 portion of an amino acid sequence as described in embodiment 24.
  • Embodiment 36.
  • A nucleic acid sequence that encodes a polypeptide as described in embodiment 35.
  • Embodiment 37.
  • A polypeptide comprising the CDR2 portion of an amino acid sequence as described in embodiment 24.
  • Embodiment 38.
  • A nucleic acid sequence that encodes a polypeptide as described in embodiment 37.
  • Embodiment 39.
  • A polypeptide comprising the CDR1 portion of an amino acid sequence as described in embodiment 24.
  • Embodiment 40.
  • A nucleic acid sequence that encodes a polypeptide as described in embodiment 39.
  • Embodiment 41.
  • A VH antibody fragment comprising at least one amino acid sequence selected from the group consisting of: SEQ lD NO:8-22.
  • Embodiment 42.
  • A VL antibody fragment comprising at least one amino acid sequence selected from the group consisting of: SEQ lD NO:23-54.
  • Embodiment 43.
  • A nucleic acid sequence that encodes a VH or VL antibody fragment as described in embodiments 41 or 42.
  • Embodiment 44.
  • A VH antibody fragment comprising at least two amino acid sequences as described in embodiment 41.
  • Embodiment 45.
  • A VL antibody fragment comprising at least two amino acid sequences as described in embodiment 42.
  • Embodiment 46.
  • A nucleic acid sequence that encodes a VH or VL antibody fragment as described in embodiments 44 or 45.
  • Embodiment 47.
  • A VH antibody fragment comprising at least three amino acid sequences as described in embodiment 41.
  • Embodiment 48.
  • A VL antibody fragment comprising at least three amino acid sequences as described in embodiment 42.
  • Embodiment 49.
  • A nucleic acid sequence that encodes a VH or VL antibody fragment as described in embodiment 47 or 48.
  • Embodiment 50.
  • A multimer comprising at least two VH antibody fragments as described in embodiment 41, 44 or 47.
  • Embodiment 51.
  • A multimer comprising at least two VL antibody fragments as described in embodiments 42, 45 or 48.
  • Embodiment 52.
  • A multimer comprising at least one VH antibody fragment as described in embodiments 41, 44 or 47, and at least one VL antibody fragment as described in embodiments 42, 45 or 48.
  • Embodiment 53.
  • A dimer comprising two VH antibody fragments as described in embodiment 41, 44 or 47.
  • Embodiment 54.
  • A dimer comprising two VL antibody fragments as described in embodiment 42, 45 or 48.
  • Embodiment 55.
  • A trimer comprising three VH antibody fragments as described in embodiment 41, 44 or 47.
  • Embodiment 56.
  • A trimer comprising three VL antibody fragments as described in embodiments 42, 45 or 48.
  • Embodiment 57.
  • A pentamer comprising five VH antibody fragments as described in embodiments 41, 44 or 47.
  • Embodiment 58.
  • A pentamer comprising five VL antibody fragments as described in embodiment 42, 45 or 48.
  • Embodiment 59.
  • A VH antibody fragment identified by the method described in embodiment 1.
  • Embodiment 60.
  • A VH antibody fragment as described in embodiment 59 that is of human origin.
  • Embodiment 61.
  • A VH antibody fragment as described in embodiment 59 having an amino acid sequence that deviates from the corresponding parental germline sequence as shown in Table 1.
  • Embodiment 62.
  • A VH antibody fragment as described in embodiment 59 belonging to the V H3 family.
  • Embodiment 63.
  • A VH antibody fragment as described in embodiment 59 belonging to a V germline selected from the group consisting of DP47, V3-49, DP- 38, V3-53, YAC-5 and 8-1 B.
  • Embodiment 64.
  • A VH antibody fragment as described in embodiment 59 belonging to a J germline selected from the group consisting of JH4b, JH6c, JH3b, JH4, JH3a, and JH1.
  • Embodiment 65.
  • A VH antibody fragment as described in embodiment 59 and having a residue other than valine at position 37 of its amino acid sequence.
  • Embodiment 66.
  • A VH antibody fragment as described in embodiment 63 having a phenylalanine residue at position 37 of its amino acid sequence.
  • Embodiment 67.
  • A VH antibody fragment as described in embodiment 63 having a tyrosine residue at position 37 of its amino acid sequence.
  • Embodiment 68.
  • A VH antibody fragment as described in embodiment 59 having a mutation at a position selected from the group consisting of positions 1,5,6,35,83,84,84a and 108 of its amino acid sequence.
  • Embodiment 69.
  • A VH antibody fragment as described in embodiment 59 which binds to protein A.
  • Embodiment 70.
  • A VL antibody fragment identified by the described in embodiment 1.
  • Embodiment 71.
  • A VL antibody fragment as described in embodiment 70 that is of human origin.
  • Embodiment 72.
  • A VL antibody fragment as described in embodiment 70 belonging to subgroup V K1 , V K3 or Vλ1.
  • Embodiment 73.
  • A VL antibody fragment as described in embodiment 70 belonging to the kappa class.
  • Embodiment 74.
  • A VL antibody fragment as described in embodiment 70 belonging to a V germline selected from the group consisting of L6, A27, L2, L16,02/012, A30 and 1 b.
  • Embodiment 75.
  • A VL antibody fragment as described in embodiment 70 belonging to a J germline selected from the group consisting of J K1, J K4, J K2, and Jλ3b.
  • Embodiment 76.
  • A VL antibody fragment as described in embodiment 70 having an arginine, proline, lysine, threonine, leucine, serine, tyrosine, glutamic acid, glutamine or histidine residue at position 96 of its amino acid sequence.
  • Embodiment 77.
  • A VL antibody fragment as described in embodiment 70 having a charged amino acid residue at position 96 of its amino acid sequence.
  • Embodiment 78.
  • A VL antibody fragment as described in embodiment 70 which binds to protein L.
  • Embodiment 79.
  • A VL antibody fragment as described in embodiment 70 having a threonine, valine or leucine amino acid residue at position 105, 106 and 107 of its amino acid sequence.
  • Embodiment 80.
  • A polypeptide sequence identified by the method described in embodiment 1.
  • Embodiment 81.
  • A nucleic acid sequence that encodes the polypeptide sequence described in embodiment 80.
  • Embodiment 82.
  • A display library constructed using polypeptide sequences as described in embodiment 80.
  • Embodiment 83.
  • A display library constructed using polypeptide sequences as described in embodiments 24, 59 or 70.
  • Embodiment 84.
  • A display library constructed using VH or VL antibody fragments as described in embodiments 41, 42, 44, 45, 47 or 48.
  • Embodiment 85.
  • A display library constructed using polypeptide sequences as described in embodiments 27, 37 or 39.
  • Embodiment 86.
  • A display library constructed using the loop (L1, L2, L3, H1, H2, or H3) regions of the polypeptides described in embodiment 24, 59 or 70.
  • Embodiment 87.
  • A display library as described in embodiment 82, 83, 84, 85 or 86, that is a phage display library.
  • Embodiment 88.
  • A display library as described in embodiment 82, 83, 84, 85 or 86 that is a ribosome display, ARM ribosome display, yeast display, bacterial cell display or in vitro compartmentalization library.
  • Embodiment 89.
  • A method for producing polypeptides with desirable biophysical properties, comprising the steps of a) providing at least one first nucleic acid sequence that encodes an antibody fragment as described in embodiment 41,42,44,45,47,48, 59 or 70 or that encodes a polypeptide sequence described in embodiment 24,27,37 or 39, and having a first desirable property; b) providing at least one second nucleic acid sequence that encodes an antibody fragment having a second desirable property; c) cutting the at least one first and at least one second nucleic acid sequences into random fragments; d) reassembling the random fragments; e) expressing the random fragments; and f) screening the expressed random fragments for the first and second desirable properties.
  • Embodiment 90.
  • A method as described in embodiment 89, wherein the first desirable property is selected from the group consisting of solubility, stability, monomericity, high expression, human origin, and binding specificity.
  • Embodiment 91.
  • A method as described in embodiment 89, wherein the second desirable property is selected from the group consisting of solubility, stability, monomericity, high expression, human origin and binding specificity.
  • Embodiment 92.
  • A method as described in embodiment 89, where the at least one second antibody fragment is of non-human origin.
  • Embodiment 93.
  • A method as described in embodiment 89, where the at least one second antibody fragment is of human origin.
  • Embodiment 94.
  • A method as described in embodiment 89, where the at least one second antibody fragment is a human VH or VL.
  • Embodiment 95.
  • A method as described in embodiment 89, where screening of the reassembled random fragments is accomplished by ribosome display, yeast display, phage display, bacterial cell display, ARM ribosome display or in vitro compartmentalization.
  • Embodiment 96.
  • A pharmaceutical composition comprising the antibody fragment described in embodiment 59 or 70, and a pharmaceutically suitable agent.
  • Embodiment 97.
  • A pharmaceutical composition comprising the polypeptide sequence described in embodiment 24 or 80, and a pharmaceutically suitable agent.
  • Embodiment 98.
  • A recombinant vector comprising the nucleic acid sequence described in embodiment 26 or 81.
  • Embodiment 99.
  • A host cell transformed with the recombinant vector described in embodiment 98.
    Figure imgb0002
    Figure imgb0003
    Figure imgb0004
    Figure imgb0005
    Figure imgb0006
    Figure imgb0007
    Figure imgb0008
    Figure imgb0009
    Figure imgb0010
    Figure imgb0011
    Figure imgb0012
    Figure imgb0013
    Figure imgb0014
    Figure imgb0015
    Figure imgb0016
    Figure imgb0017
    Figure imgb0018
    Figure imgb0019
    Figure imgb0020
    Figure imgb0021
    Figure imgb0022
    Figure imgb0023
    Figure imgb0024
    Figure imgb0025
    Figure imgb0026
    Figure imgb0027
    Figure imgb0028

Claims (15)

  1. An antibody fragment comprising a framework region 1 (FR1) sequence of ETTLTQSPATLSVSPGERATFSC, a FR2 sequence of WYQQKPGQAPRLLlY, a FR3 sequence of GlPDRFSASGSGTDFTLTlSRLEPEDFAVYYC, and a FR4 sequence of FGQGTKVElK.
  2. An antibody fragment comprising the FR1, FR2, FR3, and FR4 portion of the sequence of SEQ lD NO:46.
  3. A nucleic acid sequence encoding the antibody fragment of claim 1.
  4. A multimer comprising the antibody fragment of claim 1.
  5. A dimer comprising the antibody fragment of claim 1.
  6. A trimer comprising the antibody fragment of claim 1.
  7. A pentamer comprising the antibody fragment of claim 1.
  8. A display library constructed based on the antibody fragment sequence of claim 1.
  9. The display library of claim 8, wherein the library is a phage display library.
  10. The display library of claim 8, wherein the library is a ribosome display, ARM ribosome display, yeast display, bacterial cell display, or in vitro compartmentalization library.
  11. A method for producing an antibody fragment library, comprising:
    a) providing a nucleotide sequence encoding the antibody fragment of claim 1;
    b) providing oligonucleotide sequences with randomized codons;
    c) incorporating the randomized oligonucleotides into the nucleotide sequence encoding the antibody fragment, such that one or more than one of the complementary determining regions is randomized;
    d) expressing the nucleotide sequences produced in step c); and
    e) screening the expressed sequences for binding to a target polypeptide.
  12. A recombinant vector comprising the nucleic acid sequence of claim 3.
  13. A host cell transformed with the recombinant vector of claim 10.
  14. A pharmaceutical composition comprising the antibody fragment of claim 1, and a pharmaceutically suitable agent.
  15. A method of screening the display library of claim 8, comprising expressing the antibody fragment and panning against a target molecule.
EP10190246A 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides Withdrawn EP2333067A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US66495405P 2005-03-25 2005-03-25
EP06721715A EP1869182B1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
EP06721715.8 Division 2006-03-24

Publications (1)

Publication Number Publication Date
EP2333067A1 true EP2333067A1 (en) 2011-06-15

Family

ID=37023360

Family Applications (46)

Application Number Title Priority Date Filing Date
EP10190153A Withdrawn EP2322622A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190091A Withdrawn EP2338999A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190180A Withdrawn EP2345723A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190143.7A Active EP2374886B1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190236A Withdrawn EP2336327A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190177A Withdrawn EP2339000A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190102A Withdrawn EP2368993A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190082A Withdrawn EP2316946A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190237A Withdrawn EP2336328A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190225.2A Active EP2377935B1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190235A Withdrawn EP2371961A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190147A Withdrawn EP2368995A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190162A Withdrawn EP2322623A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190240A Withdrawn EP2377936A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190086A Withdrawn EP2368990A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190205A Withdrawn EP2338997A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190232A Withdrawn EP2316945A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190173A Withdrawn EP2371960A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190242.7A Active EP2360256B1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190246A Withdrawn EP2333067A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190104A Withdrawn EP2336323A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP06721715A Not-in-force EP1869182B1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190222A Withdrawn EP2316948A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190089A Withdrawn EP2333068A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190145A Withdrawn EP2368994A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190155.1A Not-in-force EP2360255B1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190100A Withdrawn EP2368992A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190169A Withdrawn EP2336326A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190249A Withdrawn EP2338998A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190201A Withdrawn EP2374887A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190248.4A Not-in-force EP2333066B1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190194A Withdrawn EP2322621A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190215A Withdrawn EP2330195A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190093.4A Not-in-force EP2368991B1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190165A Withdrawn EP2336325A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190160A Withdrawn EP2316947A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190149A Withdrawn EP2363485A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190150A Withdrawn EP2336324A2 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190210A Withdrawn EP2351842A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190247A Withdrawn EP2330196A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190158A Withdrawn EP2339001A2 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190187.4A Not-in-force EP2322624B1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190176A Withdrawn EP2351841A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190206A Withdrawn EP2386638A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190244A Withdrawn EP2368997A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190233A Withdrawn EP2368996A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides

Family Applications Before (19)

Application Number Title Priority Date Filing Date
EP10190153A Withdrawn EP2322622A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190091A Withdrawn EP2338999A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190180A Withdrawn EP2345723A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190143.7A Active EP2374886B1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190236A Withdrawn EP2336327A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190177A Withdrawn EP2339000A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190102A Withdrawn EP2368993A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190082A Withdrawn EP2316946A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190237A Withdrawn EP2336328A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190225.2A Active EP2377935B1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190235A Withdrawn EP2371961A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190147A Withdrawn EP2368995A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190162A Withdrawn EP2322623A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190240A Withdrawn EP2377936A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190086A Withdrawn EP2368990A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190205A Withdrawn EP2338997A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190232A Withdrawn EP2316945A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190173A Withdrawn EP2371960A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190242.7A Active EP2360256B1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides

Family Applications After (26)

Application Number Title Priority Date Filing Date
EP10190104A Withdrawn EP2336323A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP06721715A Not-in-force EP1869182B1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190222A Withdrawn EP2316948A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190089A Withdrawn EP2333068A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190145A Withdrawn EP2368994A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190155.1A Not-in-force EP2360255B1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190100A Withdrawn EP2368992A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190169A Withdrawn EP2336326A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190249A Withdrawn EP2338998A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190201A Withdrawn EP2374887A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190248.4A Not-in-force EP2333066B1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190194A Withdrawn EP2322621A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190215A Withdrawn EP2330195A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190093.4A Not-in-force EP2368991B1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190165A Withdrawn EP2336325A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190160A Withdrawn EP2316947A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190149A Withdrawn EP2363485A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190150A Withdrawn EP2336324A2 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190210A Withdrawn EP2351842A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190247A Withdrawn EP2330196A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190158A Withdrawn EP2339001A2 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190187.4A Not-in-force EP2322624B1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190176A Withdrawn EP2351841A1 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190206A Withdrawn EP2386638A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190244A Withdrawn EP2368997A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides
EP10190233A Withdrawn EP2368996A3 (en) 2005-03-25 2006-03-24 Method for isolation of soluble polypeptides

Country Status (10)

Country Link
US (5) US8293233B2 (en)
EP (46) EP2322622A3 (en)
JP (7) JP5171611B2 (en)
AT (1) ATE531800T1 (en)
AU (1) AU2006227536B9 (en)
CA (8) CA2868867A1 (en)
ES (8) ES2531537T3 (en)
PL (1) PL2360255T3 (en)
PT (1) PT2360255E (en)
WO (1) WO2006099747A1 (en)

Families Citing this family (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2278013A1 (en) 2001-04-16 2011-01-26 Wyeth Holdings Corporation Streptococcus pneumoniae open reading frames encoding polypeptide antigens and uses thereof
CN101175508A (en) 2004-10-21 2008-05-07 惠氏公司 Immunogenic compositions of staphylococcus epidermidis polypeptide antigens
EP2322622A3 (en) 2005-03-25 2012-03-07 National Research Council of Canada Method for isolation of soluble polypeptides
CA2689941C (en) 2007-06-25 2019-10-29 Esbatech Ag Methods of modifying antibodies, and modified antibodies with improved functional properties
US9181327B2 (en) * 2008-01-07 2015-11-10 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Anti-HIV domain antibodies and method of making and using same
US20110269148A1 (en) * 2008-11-26 2011-11-03 National Cheng Chung University Nanoaggregate Embedded Beads Conjugated To Single Domain Antibodies
CA2815888C (en) * 2010-10-25 2020-06-30 National Research Council Of Canada Clostridium difficile-specific antibodies and uses thereof
EA201391086A1 (en) 2011-01-28 2014-01-30 Нэшнл Рисеч Каунсил Оф Канада CONSTRUCTION OF IMMUNOGLOBULIN DOMAINS
WO2014043509A2 (en) * 2012-09-13 2014-03-20 Novartis Ag Antigen binding molecule with terminal modifications
DK2989203T3 (en) 2013-04-23 2020-02-17 The Univ Court Of The Univ Of Aberdeen SYNTHETIC LIBRARY FOR SPECIFIC BINDING MOLECULES
US10184008B2 (en) 2014-12-19 2019-01-22 Medimmune Limited Blood brain barrier transport molecules and uses thereof
RU2689689C2 (en) 2015-01-23 2019-05-28 Хеликс Байофарма Корпорейшн Conjugates of antibody-urease for therapeutic purposes
CN108697791B (en) * 2015-11-03 2022-08-23 詹森生物科技公司 Antibodies that specifically bind to PD-1 and uses thereof
KR20200128703A (en) * 2018-03-06 2020-11-16 프레시전 인코포레이티드 Hepatitis B vaccine and uses thereof
JP2021514667A (en) * 2018-03-06 2021-06-17 プレシゲン,インコーポレイテッド Human papillomavirus vaccine and its use
WO2020041360A1 (en) 2018-08-21 2020-02-27 Quidel Corporation Dbpa antibodies and uses thereof
CN115298213A (en) 2019-12-19 2022-11-04 奎多公司 Monoclonal antibody fusion
AR124914A1 (en) * 2021-02-18 2023-05-17 Mitsubishi Tanabe Pharma Corp NEW ANTI-PAD4 ANTIBODY

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998001581A1 (en) * 1996-07-09 1998-01-15 Recombinant Biocatalysis, Inc. Method of dna shuffling with polynucleotides produced by blockingor interrupting a synthesis or amplification process
WO2002051870A2 (en) * 2000-12-22 2002-07-04 GRAD, Carole Legal Representative of KAPLAN, Howard Phage display libraries of human vh fragments
WO2002059340A1 (en) * 2001-01-26 2002-08-01 The Scripps Research Institute Immunopolypeptides to hepatitis c virus
EP1479694A2 (en) * 1999-12-28 2004-11-24 Esbatech AG Intrabodies ScFv with defined framework that is stable in a reducing environment

Family Cites Families (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5530101A (en) * 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
WO1992002551A1 (en) * 1990-08-02 1992-02-20 B.R. Centre Limited Methods for the production of proteins with a desired function
CA2103887C (en) * 1991-12-13 2005-08-30 Gary M. Studnicka Methods and materials for preparation of modified antibody variable domains and therapeutic uses thereof
US6596850B1 (en) 1998-01-30 2003-07-22 Ixsys, Incorporated Anti-αv3β3 recombinant human antibodies, nucleic acids encoding same
US5994511A (en) * 1997-07-02 1999-11-30 Genentech, Inc. Anti-IgE antibodies and methods of improving polypeptides
JP4540856B2 (en) * 1999-04-09 2010-09-08 ウニヴェルジテート・チューリッヒ Novel method for stabilizing chimeric immunoglobulins or immunoglobulin fragments and stabilized anti-EGP-2 scFv fragments
GB9911569D0 (en) * 1999-05-18 1999-07-21 Oxford Biomedica Ltd Antibodies
IL151972A0 (en) * 2000-03-27 2003-04-10 Univ California Ligands directed to the non-secretory component, non-stalk region of pigr and methods of use thereof
WO2001072771A2 (en) * 2000-03-29 2001-10-04 Dgi Biotechnologies, L.L.C. Insulin and igf-1 receptor agonists and antagonists
ATE365749T1 (en) 2000-05-12 2007-07-15 Gpc Biotech Ag HUMAN PEPTIDES/PROTEINS THAT CAUSE OR EFFECT THE KILLING OF CELLS, INCLUDING LYMPHOID TUMOR CELLS
WO2002002641A1 (en) * 2000-06-16 2002-01-10 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to blys
JP3375941B2 (en) * 2000-11-24 2003-02-10 科学技術振興事業団 Highly efficient antibody screening method
DE10123041A1 (en) * 2001-05-11 2002-11-28 Tobias Heintges New single-chain human antibody fragment, useful for treating or diagnosing hepatitis C virus infection, has affinity for an essential viral protein
WO2003014960A2 (en) * 2001-08-03 2003-02-20 Medical Research Council Method of identifying a consensus sequence for intracellular antibodies
WO2003019187A1 (en) * 2001-08-22 2003-03-06 Institute For Antibodies Co., Ltd Method of selecting binding molecule
GB0124317D0 (en) * 2001-10-10 2001-11-28 Celltech R&D Ltd Biological products
WO2005016236A2 (en) * 2003-05-06 2005-02-24 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to trail receptors
EP1485075A4 (en) 2002-02-20 2006-04-26 Dyax Corp Mhc-peptide complex binding ligands
GB0226727D0 (en) 2002-11-15 2002-12-24 Medical Res Council Intrabodies
AU2003238370B2 (en) 2002-05-22 2010-04-29 Novartis Ag Immunoglobulin frameworks which demonstrate enhanced stability in the intracellular environment and methods of identifying same
AU2003240822A1 (en) * 2002-05-30 2003-12-19 Human Genome Sciences, Inc. Antibodies that specifically bind to neurokinin b
WO2003104425A2 (en) 2002-06-07 2003-12-18 The Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services Novel stable anti-cd22 antibodies
JP2006512895A (en) * 2002-06-28 2006-04-20 ドマンティス リミテッド Ligand
AU2003251791B2 (en) * 2002-07-03 2009-07-23 The Trustees Of The University Of Pennsylvania Compositions, methods and kits relating to anti-platelet autoantibodies and inhibitors thereof
AU2003282247A1 (en) * 2002-11-15 2004-06-15 Medical Research Council Anti-activated ras antibodies
ES2340280T3 (en) * 2003-03-14 2010-06-01 Wyeth Llc DIRECTED ANTIBODIES AGAINST THE RECEIVER OF HUMAN IL-21 AND USE OF THE SAME.
AR045614A1 (en) 2003-09-10 2005-11-02 Hoffmann La Roche ANTIBODIES AGAINST THE RECEIVER OF INTERLEUQUINA- 1 AND USES OF THE SAME
TWI333977B (en) * 2003-09-18 2010-12-01 Symphogen As Method for linking sequences of interest
CA2560889A1 (en) 2004-03-23 2005-10-13 Sanjay D. Khare Monoclonal antibodies specific for human ox40l (cd 134l)
PT1773885E (en) * 2004-08-05 2010-07-21 Genentech Inc Humanized anti-cmet antagonists
JP2008532476A (en) * 2004-08-12 2008-08-21 ダイアックス コーポレーション Complex binding protein
EP2322622A3 (en) * 2005-03-25 2012-03-07 National Research Council of Canada Method for isolation of soluble polypeptides
US20110269148A1 (en) * 2008-11-26 2011-11-03 National Cheng Chung University Nanoaggregate Embedded Beads Conjugated To Single Domain Antibodies
EA201391086A1 (en) * 2011-01-28 2014-01-30 Нэшнл Рисеч Каунсил Оф Канада CONSTRUCTION OF IMMUNOGLOBULIN DOMAINS
WO2014043509A2 (en) * 2012-09-13 2014-03-20 Novartis Ag Antigen binding molecule with terminal modifications

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998001581A1 (en) * 1996-07-09 1998-01-15 Recombinant Biocatalysis, Inc. Method of dna shuffling with polynucleotides produced by blockingor interrupting a synthesis or amplification process
EP1479694A2 (en) * 1999-12-28 2004-11-24 Esbatech AG Intrabodies ScFv with defined framework that is stable in a reducing environment
WO2002051870A2 (en) * 2000-12-22 2002-07-04 GRAD, Carole Legal Representative of KAPLAN, Howard Phage display libraries of human vh fragments
WO2002059340A1 (en) * 2001-01-26 2002-08-01 The Scripps Research Institute Immunopolypeptides to hepatitis c virus

Non-Patent Citations (29)

* Cited by examiner, † Cited by third party
Title
BAI, Y.; FENG, H.: "Selection of stably folded proteins by phage-display with proteolysis", EUR.J.BIOCHEM., vol. 271, 2004, pages 1609 - 1614
COX J P L ET AL: "A directory of human germ-line Vkappa segments reveals a strong bias in their usage", EUROPEAN JOURNAL OF IMMUNOLOGY, WILEY - V C H VERLAG GMBH & CO. KGAA, DE, vol. 24, 1 January 1994 (1994-01-01), pages 827 - 836, XP003004702, ISSN: 0014-2980, DOI: 10.1002/EJI.1830240409 *
DAVIES, J.; RIECHMANN, L.: "Antibody VH domains as small recognition units", BIOTECHNOLOGY N.Y, vol. 13, 1995, pages 475 - 479
DAVIES, J.; RIECHMANN, L.: "Camelising' human antibody fragments: NMR studies on VH domains", FEBS LETT, vol. 339, 21 February 1994 (1994-02-21), pages 285 - 290
DE HAARD, H. J.; VAN NEER, N.; REURS, A.; HUFTON, S. E.; ROOVERS, R. C.; HENDERIKX, P.; DE BRUINE, A. P.; ARENDS, J. W.; HOOGENBOO: "A large non-immunized human Fab fragment phage library that permits rapid isolation and kinetic analysis of high affinity antibodies", J.BIOL.CHEM., vol. 274, 25 June 1999 (1999-06-25), pages 18218 - 18230
DENG, S. J.; MACKENZIE, C. R.; HIRAMA, T.; BROUSSEAU, R.; LOWARY, T. L.; YOUNG, N. M.; BUNDLE, D. R.; NARANG, S. A.: "Basis for selection of improved carbohydrate-binding single-chain antibodies from synthetic gene libraries", PROC.NATL.ACAD.SCI U.S.A, vol. 92, 23 May 1995 (1995-05-23), pages 4992 - 4996
FORRER, P.; JUNG, S.; PLUCKTHUN, A.: "Beyond binding: using phage display to select for structure, folding and enzymatic activity in proteins", CURR.OPIN.STRUCT.BIOL., vol. 9, 1999, pages 514 - 520
FOURNIER, B.; KLIER, A.: "Protein A gene expression is regulated by DNA supercoiling which is modified by the ArIS-ArIR two-component system of Staphylococcus aureus", MICROBIOLOGY, vol. 150, 2004, pages 3807 - 3819
HAMERS, C. C.; ATARHOUCH, T.; MUYLDERMANS, S.; ROBINSON, G.; HAMERS, C.; SONGA, E. B.; BENDAHMAN, N.; HAMERS, R.: "Naturally occurring antibodies devoid of light chains", NATURE, vol. 363, 3 June 1993 (1993-06-03), pages 446 - 448
HUDSON P J ET AL: "High avidity scFv multimers; diabodies and triabodies", JOURNAL OF IMMUNOLOGICAL METHODS, ELSEVIER SCIENCE PUBLISHERS B.V.,AMSTERDAM, NL, vol. 231, no. 1-2, 10 December 1999 (1999-12-10), pages 177 - 189, XP004186084, ISSN: 0022-1759, DOI: 10.1016/S0022-1759(99)00157-X *
JESPERS, L.; SCHON, O.; FAMM, K.; WINTER, G.: "Aggregation-resistant domain antibodies selected on phage by heat denaturation", NAT.BIOTECHNOL., vol. 22, 2004, pages 1161 - 1165
JESPERS, L.; SCHON, O.; JAMES, L. C.; VEPRINTSEV, D.; WINTER, G.: "Crystal Structure of HEL4, a Soluble, Refoldable Human V(H) Single Domain with a Germ-line Scaffold", J.MOL.BIOL., vol. 337, 2 April 2004 (2004-04-02), pages 893 - 903
JUNG, S.; HONEGGER, A.; PLUCKTHUN, A.: "Selection for improved protein stability by phage display", J.MOL.BIOL., vol. 294, 19 November 1999 (1999-11-19), pages 163 - 180
MATSUURA, T.; PLUCKTHUN, A.: "Selection based on the folding properties of proteins with ribosome display", FEBS LETT., vol. 539, 27 March 2003 (2003-03-27), pages 24 - 28
MURUGANANDAM, A.; TANHA, J.; NARANG, S.; STANIMIROVIC, D.: "Selection of phage-displayed llama single-domain antibodies that transmigrate across human blood-brain barrier endothelium", FASEB J., vol. 16, 2002, pages 240 - 242
PACE, C. N.; VAJDOS, F.; FEE, L.; GRIMSLEY, G.; GRAY, T.: "How to measure and predict the molar absorption coefficient of a protein", PROTEIN SCI., vol. 4, 1995, pages 2411 - 2423
RICCI, S.; MEDAGLINI, D.; MARCOTTE, H.; OLSEN, A.; POZZI, G.; BJORCK, L.: "Immunoglobulin-binding domains of pep to streptococcal protein L enhance vaginal colonization of mice by Streptococcus gordonii", MICROB.PATHOG., vol. 30, 2001, pages 229 - 235
SAMBROOK, J. F. E. F.: "Molecular Cloning: A laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY
SBLATTERO, D.; BRADBURY, A.: "A definitive set of oligonucleotide primers for amplifying human V regions", IMMUNOTECHNOLOGY, vol. 3, 1998, pages 271 - 278
TANHA, J.; DUBUC, G.; HIRAMA, T.; NARANG, S. A.; MACKENZIE, C. R.: "Selection by phage display of llama conventional V(H) fragments with heavy chain antibody V(H)H properties", J.IMMUNOL.METHODS, vol. 263, 1 May 2002 (2002-05-01), pages 97 - 109
TANHA, J.; MURUGANANDAM, A.; STANIMIROVIC, D.: "Phage Display Technology for Identifying Specific Antigens on Brain Endothelial Cells", METHODS MOL.MED., vol. 89, 2003, pages 435 - 450
TANHA, J.; XU, P.; CHEN, Z. G.; NI, F.; KAPLAN, H.; NARANG, S. A.; MACKENZIE, C. R.: "Optimal design features ofcamelized human single-domain antibody libraries", J.BIOL.CHEM, vol. 276, 7 June 2001 (2001-06-07), pages 24774 - 24780
TO R ET AL: "Isolation of Monomeric Human VHS by a Phage Selection", JOURNAL OF BIOLOGICAL CHEMISTRY, THE AMERICAN SOCIETY OF BIOLOGICAL CHEMISTS, INC, US, vol. 280, no. 50, 16 December 2005 (2005-12-16), pages 41395 - 41403, XP009100333, ISSN: 0021-9258, DOI: 10.1074/JBC.M509900200 *
VAN DER LINDEN, R. H.; FRENKEN, L. G.; DE GEUS, B.; HANNSEN, M. M.; RUULS, R. C.; STOK, W.; DE RON, L.; WILSON, S.; DAVIS, P.; VER: "Comparison of physical chemical properties of llama VHH antibody fragments and mouse monoclonal antibodies", BIOCHIM.BIOPHYS.ACTA, vol. 1431, 12 April 1999 (1999-04-12), pages 37 - 46
WALDO, G. S.: "Genetic screens and directed evolution for protein solubility", CURR.OPIN.CHEM.BIOL., vol. 7, 2003, pages 33 - 38
WANG XIANG-BIN ET AL: "A new approach for rapidly reshaping single-chain antibody in vitro by combining DNA shuffling with ribosome display", JOURNAL OF BIOCHEMISTRY (TOKYO), vol. 136, no. 1, July 2004 (2004-07-01), pages 19 - 28, XP002626886, ISSN: 0021-924X *
WARD, E. S.; GUSSOW, D.; GRIFFITHS, A. D.; JONES, P. T.; WINTER, G.: "Binding activities of a repertoire of single immunoglobulin variable domains secreted from Escherichia coli [see comments]", NATURE, vol. 341, 12 October 1989 (1989-10-12), pages 544 - 546
WOERN A ET AL: "Stability engineering of antibody single-chain Fv fragments", JOURNAL OF MOLECULAR BIOLOGY, LONDON, GB, vol. 305, no. 5, 1 January 2001 (2001-01-01), pages 989 - 1010, XP002249286, ISSN: 0022-2836, DOI: 10.1006/JMBI.2000.4265 *
ZHANG, J.; LI, Q.; NGUYEN, T. D.; TREMBLAY, T. L.; STONE, E.; TO, R.; KELLY, J.; MACKENZIE, C. R.: "A pentavalent single-domain antibody approach to tumor antigen discovery and the development of novel proteomics reagents", J.MOL.BIOL., vol. 341, 30 July 2004 (2004-07-30), pages 161 - 169

Also Published As

Publication number Publication date
JP2016103998A (en) 2016-06-09
EP2377936A3 (en) 2012-06-13
EP2371961A1 (en) 2011-10-05
CA2602028C (en) 2015-01-13
EP2371960A2 (en) 2011-10-05
JP6374851B2 (en) 2018-08-15
CA2873899A1 (en) 2006-09-28
ES2550836T3 (en) 2015-11-12
EP2363485A1 (en) 2011-09-07
JP5171611B2 (en) 2013-03-27
EP2338999A2 (en) 2011-06-29
CA2868774A1 (en) 2006-09-28
EP2368997A2 (en) 2011-09-28
EP2368996A3 (en) 2011-12-14
EP2351842A1 (en) 2011-08-03
EP2377936A2 (en) 2011-10-19
EP2368997A3 (en) 2012-02-22
EP2360255A1 (en) 2011-08-24
ES2558338T3 (en) 2016-02-03
EP2322624A2 (en) 2011-05-18
EP2368994A3 (en) 2012-04-18
EP2374887A2 (en) 2011-10-12
EP2316948A1 (en) 2011-05-04
EP2316946A1 (en) 2011-05-04
EP2368990A1 (en) 2011-09-28
EP2338999A3 (en) 2012-05-23
EP2368995A2 (en) 2011-09-28
EP2322621A3 (en) 2012-09-12
EP2377935A1 (en) 2011-10-19
EP2368992A2 (en) 2011-09-28
EP2322624A3 (en) 2012-03-14
EP2374886A3 (en) 2011-11-23
EP2351841A1 (en) 2011-08-03
EP2336324A2 (en) 2011-06-22
EP2322623A3 (en) 2011-10-19
CA2602028A1 (en) 2006-09-28
PT2360255E (en) 2015-04-08
JP2014144010A (en) 2014-08-14
CA2873906A1 (en) 2006-09-28
JP6628822B2 (en) 2020-01-15
EP2374887A3 (en) 2012-07-18
EP2322622A3 (en) 2012-03-07
EP2322623A2 (en) 2011-05-18
EP2336326A3 (en) 2012-02-15
ES2531537T3 (en) 2015-03-17
EP2316947A1 (en) 2011-05-04
EP2330196A2 (en) 2011-06-08
US11091536B2 (en) 2021-08-17
EP2338997A1 (en) 2011-06-29
EP2333066A1 (en) 2011-06-15
EP2336326A2 (en) 2011-06-22
EP2377935B1 (en) 2015-07-15
EP2368993A2 (en) 2011-09-28
AU2006227536A1 (en) 2006-09-28
JP2016073284A (en) 2016-05-12
EP1869182A4 (en) 2008-08-13
EP2322622A2 (en) 2011-05-18
EP2339001A2 (en) 2011-06-29
EP2374886B1 (en) 2015-11-11
EP2345723A1 (en) 2011-07-20
EP2360256A1 (en) 2011-08-24
WO2006099747A1 (en) 2006-09-28
EP2339000A1 (en) 2011-06-29
ES2546788T3 (en) 2015-09-28
EP2322621A2 (en) 2011-05-18
US20160052995A1 (en) 2016-02-25
EP2368991B1 (en) 2016-02-17
ES2375826T3 (en) 2012-03-06
US8293233B2 (en) 2012-10-23
JP2018110583A (en) 2018-07-19
US20090220485A1 (en) 2009-09-03
ES2565782T3 (en) 2016-04-06
JP2012070748A (en) 2012-04-12
EP2336323A2 (en) 2011-06-22
US20220017603A1 (en) 2022-01-20
AU2006227536B2 (en) 2011-08-11
US20130142780A1 (en) 2013-06-06
ATE531800T1 (en) 2011-11-15
CA2868867A1 (en) 2006-09-28
EP2368991A2 (en) 2011-09-28
CA2873899C (en) 2020-11-10
EP2371960A3 (en) 2012-01-11
JP5731956B2 (en) 2015-06-10
US20190092840A1 (en) 2019-03-28
EP2330195A3 (en) 2012-05-23
EP2360255B1 (en) 2015-01-28
EP2336328A1 (en) 2011-06-22
EP1869182A1 (en) 2007-12-26
EP2374886A2 (en) 2011-10-12
CA2873939C (en) 2020-11-10
EP2330196A3 (en) 2012-04-18
EP2316945A1 (en) 2011-05-04
EP2333066B1 (en) 2015-02-18
EP2338998A1 (en) 2011-06-29
EP2333068A1 (en) 2011-06-15
EP2330195A2 (en) 2011-06-08
CA2873906C (en) 2020-11-24
EP2368996A2 (en) 2011-09-28
JP2019187439A (en) 2019-10-31
CA2873939A1 (en) 2006-09-28
EP1869182B1 (en) 2011-11-02
EP2336325A1 (en) 2011-06-22
EP2368993A3 (en) 2012-06-13
EP2336323A3 (en) 2012-05-23
EP2322624B1 (en) 2015-09-23
CA2868865A1 (en) 2006-09-28
PL2360255T3 (en) 2015-05-29
JP6878508B2 (en) 2021-05-26
EP2386638A2 (en) 2011-11-16
EP2368994A2 (en) 2011-09-28
EP2386638A3 (en) 2012-08-15
JP2008536490A (en) 2008-09-11
ES2536995T3 (en) 2015-06-01
EP2336327A1 (en) 2011-06-22
JP5901679B2 (en) 2016-04-13
CA2868781A1 (en) 2006-09-28
EP2368995A3 (en) 2012-01-18
ES2534305T3 (en) 2015-04-21
EP2368991A3 (en) 2012-07-25
US10150807B2 (en) 2018-12-11
AU2006227536B9 (en) 2011-08-18
EP2360256B1 (en) 2014-12-31
EP2368992A3 (en) 2012-03-14

Similar Documents

Publication Publication Date Title
EP2368991B1 (en) Method for isolation of soluble polypeptides

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

AC Divisional application: reference to earlier application

Ref document number: 1869182

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

17P Request for examination filed

Effective date: 20111214

17Q First examination report despatched

Effective date: 20120227

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20121205